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










Publication year range
1.
Biomater Sci ; 12(15): 3933-3946, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38940612

ABSTRACT

PEGylation is currently used for the synthesis of stealth liposomes and to enhance the pharmacokinetic and biopharmaceutical properties of payloads. PEGylated dendron phospholipids can decrease the detachment of polyethylene glycol (PEG) from the liposomal surface owing to an increased hydrophobic anchoring effect on the phospholipid bilayer of liposomes and thus generating super stealth liposomes that are suitable for the systemic delivery of anticancer drugs. Herein, doxorubicin hydrochloride-loaded super stealth liposomes were studied for the treatment of breast cancer lung metastasis in an animal model. The results demonstrated that the super stealth liposomes had suitable physicochemical properties for in vivo administration and could significantly increase the efficacy of doxorubicin in breast cancer lung metastasis tumor-bearing mice compared to the free drug. The super stealth liposomes also increased doxorubicin accumulation inside the tumor tissue. The permanence of PEG on the surface of the super stealth liposomes favored the formation of a depot of therapeutic nanocarriers inside the tumor tissue by improving their permanence after stopping treatment. The doxorubicin-loaded super stealth liposomes increased the survival of the mouse tumor model. These promising results demonstrate that the doxorubicin-loaded super stealth liposomes could be an effective nanomedicine to treat metastatic breast cancer.


Subject(s)
Breast Neoplasms , Doxorubicin , Liposomes , Polyethylene Glycols , Doxorubicin/chemistry , Doxorubicin/pharmacology , Doxorubicin/administration & dosage , Doxorubicin/analogs & derivatives , Animals , Liposomes/chemistry , Polyethylene Glycols/chemistry , Polyethylene Glycols/administration & dosage , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Female , Mice , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Humans , Cell Line, Tumor , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Mice, Inbred BALB C , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacokinetics
2.
Biomacromolecules ; 24(9): 4229-4239, 2023 09 11.
Article in English | MEDLINE | ID: mdl-37638739

ABSTRACT

In this study, a circular conjugate of granulocyte colony-stimulating factor (G-CSF) was prepared by conjugating the two end-chains of poly(ethylene glycol) (PEG) to two different sites of the protein. For the orthogonal conjugation, a heterobifunctional PEG chain was designed and synthesized, bearing the dipeptide ZGln-Gly (ZQG) at one end-chain, for transglutaminase (TGase) enzymatic selective conjugation at Lys41 of G-CSF, and an aldehyde group at the opposite end-chain, for N-terminal selective reductive alkylation of the protein. The cPEG-Nter/K41-G-CSF circular conjugate was characterized by physicochemical methods and compared with native G-CSF and the corresponding linear monoconjugates of G-CSF, PEG-Nter-G-CSF, and PEG-K41-G-CSF. The results demonstrated that the circular conjugate had improved physicochemical and thermal stability, prolonged pharmacokinetic interaction, and retained the biological activity of G-CSF. The PEGylation strategy employed in this study has potential applications in the design of novel protein-based therapeutics.


Subject(s)
Aldehydes , Granulocyte Colony-Stimulating Factor , Alkylation , Chemical Phenomena , Dipeptides
3.
Adv Healthc Mater ; 12(29): e2301650, 2023 11.
Article in English | MEDLINE | ID: mdl-37590033

ABSTRACT

Liposomes play an important role in the field of drug delivery by virtue of their biocompatibility and versatility as carriers. Stealth liposomes, obtained by surface decoration with hydrophilic polyethylene glycol (PEG) molecules, represent an important turning point in liposome technology, leading to significant improvements in the pharmacokinetic profile compared to naked liposomes. Nevertheless, the generation of effective targeted liposomes-a central issue for cancer therapy-has faced several difficulties and clinical phase failures. Active targeting remains a challenge for liposomes. In this direction, a new Super Stealth Immunoliposomes (SSIL2) composed of a PEG-bi-phospholipids derivative is designed that stabilizes the polymer shielding over the liposomes. Furthermore, its counterpart, conjugated to the fragment antigen-binding of trastuzumab (Fab'TRZ -PEG-bi-phospholipids), is firmly anchored on the liposomes surface and correctly orients outward the targeting moiety. Throughout this study, the performances of SSIL2 are evaluated and compared to classic stealth liposomes and stealth immunoliposomes in vitro in a panel of cell lines and in vivo studies in zebrafish larvae and rodent models. Overall, SSIL2 shows superior in vitro and in vivo outcomes, both in terms of safety and anticancer efficacy, thus representing a step forward in targeted cancer therapy, and valuable for future development.


Subject(s)
Liposomes , Neoplasms , Animals , Liposomes/chemistry , Zebrafish , Drug Delivery Systems , Phospholipids , Polyethylene Glycols/chemistry
4.
PLoS One ; 17(3): e0265749, 2022.
Article in English | MEDLINE | ID: mdl-35316287

ABSTRACT

Ciliary neurotrophic factor (CNTF) is a neurotrophic cytokine able to induce appetite reduction, weight loss and antidiabetic effects. However, its susceptibility to neutralizing anti-CNTF antibodies in patients hampered its use for treatment of human obesity and diabetes. In addition, CNTF has a very short plasma half-life, which limits its use as a therapeutic agent. Solutions, directed to prolong its in vivo effects, vary from the implantation of encapsulated secreting cells to identification of more active variants or chemical modification of the protein itself. PEGylation is a widely used modification for shielding proteins from circulating antibodies and for increasing their plasma half-life. Here, we have selected DH-CNTF, a CNTF variant which has a 40-fold higher affinity for the CNTF receptor α accompanied by an increased activity in cellular assays. The PEGylated DH-CNTF retained the biological activity of native protein in vitro and showed a significant improvement of pharmacokinetic parameters. In an acute model of glucose tolerance, the PEG-DH-CNTF was able to reduce the glycemia in diet-induced obese animals, with a performance equaled by a 10-fold higher dose of DH-CNTF. In addition, the PEGylated DH-CNTF analog demonstrated a more potent weight loss effect than the unmodified protein, opening to the use of CNTF as weight reducing agent with treatment regimens that can better meet patient compliance thanks to reduced dosing schedules.


Subject(s)
Ciliary Neurotrophic Factor , Obesity , Animals , Ciliary Neurotrophic Factor/pharmacology , Diet , Humans , Mice , Mice, Obese , Obesity/drug therapy , Obesity/etiology , Polyethylene Glycols/pharmacology , Proteins , Receptor, Ciliary Neurotrophic Factor/metabolism , Weight Loss
5.
Mol Pharm ; 19(1): 345-353, 2022 01 03.
Article in English | MEDLINE | ID: mdl-34842438

ABSTRACT

Here, we evaluated the feasibility of non-prodrug PEG-drug conjugates to decrease the accumulation of drugs within the placental tissues. The results showed that PEG was biocompatible with the human placenta with no alteration of the basal rate of proliferation or apoptosis in term placental explants. No significant changes in the released levels of lactate dehydrogenase and the human chorionic gonadotropin were observed after PEG treatment. The cellular uptake studies revealed that conjugating Cy5.5 and haloperidol to PEG significantly reduced (by up to ∼40-fold) their uptake by the placenta. These findings highlight the viability of novel non-prodrug polymer-drug conjugates to avoid the accumulation of drugs within the placenta.


Subject(s)
Placenta/metabolism , Polyethylene Glycols/chemistry , Pregnancy Complications/drug therapy , Drug Compounding/methods , Female , Haloperidol/pharmacokinetics , Humans , Placenta/drug effects , Polyethylene Glycols/adverse effects , Polymers , Pregnancy
6.
Biomater Sci ; 10(1): 124-137, 2021 Dec 21.
Article in English | MEDLINE | ID: mdl-34796888

ABSTRACT

Three-dimensional (3D) culture systems have progressively attracted attention given their potential to overcome limitations of classical 2D in vitro systems. Among different supports for 3D cell culture, hydrogels (HGs) offer important advantages such as tunable mechanical and biological properties. Here, a biocompatible hyaluronic acid-polyethylene glycol HG was developed to explore the pro-migratory behavior of alveolar rhabdomyosarcoma (ARMS) cells. Proteomic analysis of ARMS xenografts unveiled the composition of the extracellular matrix (ECM) elucidating the most representative proteins. In parallel, HGs were obtained by the combination of a thiol-containing hyaluronic acid derivative and different polyethylene glycol (PEG) dimaleimide polymers. The selection of the optimal HG for ARMS cell growth was made based on degradation time, swelling, and cell distribution. Rheology measures and mechanical properties were assessed in the presence or absence of ECM proteins (collagen type I and fibronectin), as well as viability tests and cell distribution analysis. The role of ITGA5, the receptor of fibronectin, in determining ARMS cell migration was validated in vitro upon ITGA5 silencing. In vivo, cell dissemination and the capacity for engrafting were validated after injecting ARMS cell populations enriched for the level of ITGA5 in zebrafish embryos. To study the interactions with ARMS-specific ECM proteins (HG + P), the key players from the Rho and heat-shock pathways were investigated by reverse phase protein array (RPPA). Our data suggest that the developed 3D ARMS model is useful for identifying potential physical hallmarks that allow cancer cells to resist therapy, escape from the immune-system and increase dissemination.


Subject(s)
Hydrogels , Rhabdomyosarcoma , Animals , Cell Culture Techniques, Three Dimensional , Extracellular Matrix , Proteomics , Zebrafish
7.
Pharmaceutics ; 13(7)2021 Jun 23.
Article in English | MEDLINE | ID: mdl-34201494

ABSTRACT

Although selective tumor delivery of anticancer drugs has been sought by exploiting either passive targeting or by ligand-mediated targeting, a selective anticancer therapy remains an unmet medical need. Despite the advances which have been achieved by nanomedicines, nanosystems such as polymer-drug conjugates still miss the goal of clinical efficacy. In this study, we demonstrated that polymer-drug conjugates require a thoroughly chemical design and the right targeting agent/polymer ratio to be selective and effective towards cancer cells. In particular, two PEG conjugates carrying paclitaxel and targeted with different folic acid (FA)/PEG ratios (one or three) were investigated. The cytotoxicity study in positive (HT-29) and negative (HCT-15) FA receptor (FR)-cell lines demonstrated that the conjugates with one or three FAs were 4- or 28-fold more active in HT-29 cells, respectively. The higher activity of the 3-FA conjugate was confirmed by its strong impact on cell cycle arrest. Furthermore, FA targeting had a clear effect on migration and invasiveness of HT-29 cells, which were significantly reduced by both conjugates. Interestingly, the 3-FA conjugate showed also an improved pharmacokinetic profile in mice. The results of this study indicate that thorough investigations are needed to optimize and tune drug delivery and achieve the desired selectivity and activity towards cancer cells.

8.
Biomolecules ; 11(3)2021 03 10.
Article in English | MEDLINE | ID: mdl-33802146

ABSTRACT

In cystic fibrosis (CF), the accumulation of viscous lung secretions rich in DNA and actin is a major cause of chronic inflammation and recurrent infections leading to airway obstruction. Mucolytic therapy based on recombinant human DNase1 reduces CF mucus viscosity and promotes airway clearance. However, the marked susceptibility to actin inhibition of this enzyme prompts the research of alternative treatments that could overcome this limitation. Within the human DNase repertoire, DNase1L2 is ideally suited for this purpose because it exhibits metal-dependent endonuclease activity on plasmid DNA in a broad range of pH with acidic optimum and is minimally inhibited by actin. When tested on CF artificial mucus enriched with actin, submicromolar concentrations of DNase1L2 reduces mucus viscosity by 50% in a few seconds. Inspection of superimposed model structures of DNase1 and DNase1L2 highlights differences at the actin-binding interface that justify the increased resistance of DNase1L2 toward actin inhibition. Furthermore, a PEGylated form of the enzyme with preserved enzymatic activity was obtained, showing interesting results in terms of activity. This work represents an effort toward the exploitation of natural DNase variants as promising alternatives to DNase1 for the treatment of CF lung disease.


Subject(s)
Actins/metabolism , Cystic Fibrosis/therapy , Deoxyribonuclease I/metabolism , Deoxyribonuclease I/therapeutic use , Amino Acid Sequence , Calcium/metabolism , Catalytic Domain , Conserved Sequence , Cysteine/metabolism , DNA/isolation & purification , Deoxyribonuclease I/chemistry , Humans , Mucus , Oxidation-Reduction , Pichia/metabolism , Plasmids/isolation & purification , Polyethylene Glycols/chemistry , Protein Binding , Recombinant Proteins/isolation & purification
9.
Cell Mol Immunol ; 18(5): 1197-1210, 2021 05.
Article in English | MEDLINE | ID: mdl-33762685

ABSTRACT

One of the main goals of vaccine research is the development of adjuvants that can enhance immune responses and are both safe and biocompatible. We explored the application of the natural polymer hyaluronan (HA) as a promising immunological adjuvant for protein-based vaccines. Chemical conjugation of HA to antigens strongly increased their immunogenicity, reduced booster requirements, and allowed antigen dose sparing. HA-based bioconjugates stimulated robust and long-lasting humoral responses without the addition of other immunostimulatory compounds and proved highly efficient when compared to other adjuvants. Due to its intrinsic biocompatibility, HA allowed the exploitation of different injection routes and did not induce inflammation at the inoculation site. This polymer promoted rapid translocation of the antigen to draining lymph nodes, thus facilitating encounters with antigen-presenting cells. Overall, HA can be regarded as an effective and biocompatible adjuvant to be exploited for the design of a wide variety of vaccines.


Subject(s)
Adjuvants, Immunologic/pharmacology , Hyaluronic Acid/pharmacology , Vaccines/pharmacology , Alarmins/metabolism , Animals , Biocompatible Materials/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Female , Fluorescence , Hyaluronic Acid/chemistry , Immunity, Humoral/drug effects , Inflammation/pathology , Lymph Nodes/drug effects , Lymph Nodes/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Weight , Ovalbumin/immunology , Time Factors
10.
Article in English | MEDLINE | ID: mdl-33314717

ABSTRACT

Polymer conjugation can be considered one of the leading approaches within the vast field of nanotechnology-based drug delivery systems. In fact, such technology can be exploited for delivering an active molecule, such as a small drug, a protein, or genetic material, or it can be applied to other drug delivery systems as a strategy to improve their in vivo behavior or pharmacokinetic activities such as prolonging the half-life of a drug, conferring stealth properties, providing external stimuli responsiveness, and so on. If on the one hand, polymer conjugation with biotech drug is considered the linchpin of the protein delivery field boasting several products in clinical use, on the other, despite dedicated research, conjugation with low molecular weight drugs has not yet achieved the milestone of the first clinical approval. Some of the primary reasons for this debacle are the difficulties connected to achieving selective targeting to diseased tissue, organs, or cells, which is the main goal not only of polymer conjugation but of all delivery systems of small drugs. In light of the need to achieve better drug targeting, researchers are striving to identify more sophisticated, biocompatible delivery approaches and to open new horizons for drug targeting methodologies leading to successful clinical applications. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Toxicology and Regulatory Issues in Nanomedicine > Regulatory and Policy Issues in Nanomedicine.


Subject(s)
Drug Delivery Systems , Nanomedicine , Pharmaceutical Preparations , Polymers , Pharmaceutical Preparations/administration & dosage
11.
J Control Release ; 324: 228-237, 2020 08 10.
Article in English | MEDLINE | ID: mdl-32413454

ABSTRACT

Poly(L-glutamic acid)-co-poly(ethylene glycol) block copolymers (PLE-PEG) are here investigated as polymers for conjugation to therapeutic proteins such as granulocyte colony stimulating factor (G-CSF) and human growth hormone (hGH). PLE-PEG block copolymers are able to stabilize and protect proteins from degradation and to prolong their residence time in the blood stream, features that are made possible thanks to PEG's intrinsic properties and the simultaneous presence of the biodegradable anionic PLE moiety. When PLE-PEG copolymers are selectively tethered to the N-terminus of G-CSF and hGH, they yield homogeneous monoconjugates that preserve the protein's secondary structure. During the current study the pharmacokinetics of PLE10-PEG20k-G-CSF and PLE20-PEG20k-G-CSF derivatives and their ability to induce granulopoiesis were, respectively, assessed in Sprague-Dawley rats and in C57BL6 mice. Our results show that the bioavailability and bioactivity of the derivatives are comparable to or better than those of PEG20k-Nter-G-CSF (commercially known as Pegfilgrastim). The therapeutic effects of PLE10-PEG20k-hGH and PLE20-PEG20k-hGH derivatives tested in hypophysectomized rats demonstrate that the presence of a negatively charged PLE block enhances the biological properties of the conjugates additionally with respect to PEG20k-Nter-hGH.


Subject(s)
Glutamic Acid , Polyethylene Glycols , Animals , Mice , Mice, Inbred C57BL , Polymers , Rats , Rats, Sprague-Dawley
12.
Eur J Pharm Biopharm ; 142: 49-60, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31201855

ABSTRACT

Antibody drug conjugates (ADCs), which are obtained by coupling a potent cytotoxic agent to a monoclonal antibody (mAb), are traditionally bound in a random way to lysine or cysteine residues, with the final product's heterogeneity having an important impact on their activity, characterization, and manufacturing. A new antibody drug delivery system (ADS) based on a non-covalent linkage between a Fc-binding protein, in this case Protein A or Protein G, and a mAb was investigated in the effort to achieve greater homogeneity and to create a versatile and adaptable drug delivery system. Recombinant staphylococcal Protein A and streptococcal Protein G were chemically PEGylated at the N-terminus with a 5 kDa and a 20 kDa PEG, respectively, yielding two monoconjugates with a mass of ≈50 and ≈45 kDa. Circular dichroism studies showed that both conjugates preserved secondary structures of the protein, and isothermal titration calorimetry experiments demonstrated that their affinity for mAb was approximately 107 M-1. Upon complexation with a mAb (Trastuzumab or Rituximab), in vitro flow-cytometry analysis of the new ADSs showed high selectivity for the specific antigen expressing cells. In addition, the ADS complex based on Trastuzumab and Protein G, conjugated with a heterobifunctional 20 kDa PEG carrying the toxin Tubulysin A, had a marked cytotoxic effect on the cancer cell line overexpressing the HER2/neu receptor, thus supporting its application in cancer therapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Immunoconjugates/pharmacology , Cell Line, Tumor , Drug Delivery Systems/methods , Humans , Jurkat Cells , Receptor, ErbB-2/metabolism , Rituximab/pharmacology , Trastuzumab/pharmacology
13.
Int Rev Cell Mol Biol ; 346: 1-50, 2019.
Article in English | MEDLINE | ID: mdl-31122392

ABSTRACT

The targeted delivery of bioactive molecules to the appropriate site of action, one of the critical focuses of pharmaceutical research, improves therapeutic outcomes and increases safety at the same time; a concept envisaged by Ehrlich over 100 years ago when he described the "magic bullet" model. In the following decades, a considerable amount of research effort combined with enormous investment has carried selective drug targeting into clinical practice via the advent of monoclonal antibodies (mAbs) and antibody-drug conjugates derivatives. Additionally, a deeper understanding of physiopathological conditions of disease has permitted the tailored design of targeted drug delivery platforms that carry drugs, many copies of the same drug, and different drugs in combination to the appropriate site of action least selectively or preferentially. The acquired know-how has provided the field with the design rationale to develop a successful delivery system that will provide new and improved means to treat many intractable diseases and disorders. In this review, we discuss a wide range of molecular platforms for drug delivery, and focus on those with more success in the clinic, given their potential for targeted therapies.


Subject(s)
Drug Delivery Systems , Immunoconjugates/administration & dosage , Molecular Targeted Therapy , Animals , Antibodies, Monoclonal , Antineoplastic Agents/administration & dosage , Humans , Liposomes , Neoplasms/drug therapy
14.
J Pept Sci ; 25(4): e3155, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30809901

ABSTRACT

Previously, we reported the discovery of macrocyclic peptide triazoles (cPTs) that bind to HIV-1 Env gp120, inhibit virus cell infection with nanomolar potencies, and cause irreversible virion inactivation. Given the appealing virus-killing activity of cPTs and resistance to protease cleavage observed in vitro, we here investigated in vivo pharmacokinetics of the cPT AAR029b. AAR029b was investigated both alone and encapsulated in a PEGylated liposome formulation that was designed to slowly release inhibitor. Pharmacokinetic analysis in rats showed that the half-life of FITC-AAR029b was substantial both alone and liposome-encapsulated, 2.92 and 8.87 hours, respectively. Importantly, liposome-encapsulated FITC-AAR029b exhibited a 15-fold reduced clearance rate from serum compared with the free FITC-cPT. This work thus demonstrated both the in vivo stability of cPT alone and the extent of pharmacokinetic enhancement via liposome encapsulation. The results obtained open the way to further develop cPTs as long-acting HIV-1 inactivators against HIV-1 infection.


Subject(s)
Anti-HIV Agents/pharmacokinetics , HIV-1/drug effects , Macrocyclic Compounds/pharmacokinetics , Peptides/pharmacokinetics , Triazoles/pharmacokinetics , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Liposomes , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/pharmacology , Microbial Sensitivity Tests , Peptides/chemistry , Peptides/pharmacology , Triazoles/chemistry , Triazoles/pharmacology
15.
Adv Protein Chem Struct Biol ; 112: 123-142, 2018.
Article in English | MEDLINE | ID: mdl-29680235

ABSTRACT

Proteins hold a central role in medicine and biology, also confirmed by the several therapeutic applications based on biologic drugs. Such therapies are of great relevance thanks to high potency and safety of proteins. Nevertheless, many proteins as therapeutics might present issues like fast kidney clearance, rapid enzymatic degradation, or immunogenicity. Such defects implicate frequent administrations or administrations at high doses of the therapeutics, thus yielding or exacerbating potential side effects. A successful technology for improving the clinical profiles of proteins is the conjugation of polymers to the protein surface. The design of a protein-polymer conjugate presents critical aspects that determine the efficacy and safety of the final product. The control over stoichiometry and conjugation site is a strict criterion on which researchers have been intensively focused during the years, in order to obtain homogeneous and batch-to-batch reproducible products. An innovative site-specific conjugation strategy relies on the use of enzymes as tools to mediate polymer conjugation. Enzymatic approaches are attractive because they allow site-selective polymer conjugation at specific protein amino acids. In these reactions, the polymer is a substrate analog that replaces the native substrate. Furthermore, enzymes can count other advantages such as high yields of conversion and physiological conditions of reaction. This chapter provides a meaningful description of protein-polymer conjugation through transglutaminase-mediated and sialyltransferase-mediated enzymatic strategies, reporting the mechanism of action and some relevant examples.


Subject(s)
Polymers/metabolism , Proteins/metabolism , Sialyltransferases/metabolism , Transglutaminases/metabolism , Humans , Polymers/chemistry , Proteins/chemistry
16.
J Drug Target ; 25(9-10): 856-864, 2017.
Article in English | MEDLINE | ID: mdl-28805084

ABSTRACT

Microbial transglutaminase enzyme (mTGase) is an extremely useful enzyme that is increasingly employed in the food and pharmaceutical industries and as a tool for protein modification and tagging. The current study describes how we immobilised mTGase (iTGase) on a solid support to improve its stability during the PEGylation process by which polyethylene glycol chains are attached to protein and peptide drugs. When the enzyme was immobilised at the N-terminal sequence on agarose beads, it retained more than 53% of its starting activity. Kinetic studies on the immobilised and free mTGase disclosed a 1.7 and 1.5 fold decrease of Km and Vmax, respectively. Protein PEGylation was carried out using α-lactalbumin (α-LA) and granulocyte colony stimulating factor (G-CSF). In the former case, the iTGase showed a selective conjugation towards only one Gln residue of α-LA, avoiding formation of a mono- and bi-conjugate mixture that is achieved using the free enzyme. In the latter case, the immobilised enzyme still remained selective towards only one Gln, but avoided the undesired formation of deamidated G-CSF that took place when free mTGase was used. Overall, the results of the current study highlight the suitability of iTGase in preparing site-selective protein-polymer conjugates.


Subject(s)
Enzymes, Immobilized/analysis , Enzymes, Immobilized/metabolism , Polyethylene Glycols/metabolism , Transglutaminases/analysis , Transglutaminases/metabolism , Amino Acid Sequence , Enzyme Stability/physiology , Enzymes, Immobilized/genetics , Protein Structure, Secondary , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Transglutaminases/genetics
17.
Methods Enzymol ; 590: 317-346, 2017.
Article in English | MEDLINE | ID: mdl-28411643

ABSTRACT

PEGylation, the covalent attachment of polyethylene glycol to bioactive molecules, is one of the leading approaches used to prolong pharmacokinetics, to improve the stability, and to reduce the immunogenicity of therapeutic proteins. PEG-conjugated products are associated with better therapy outcomes and improved patient compliance. Widely applied in clinical practice, the technology is mainly used to modify proteins, peptides, and oligonucleotides but also other drug delivery systems such as the liposomal one. Undergoing continuous attempts to optimize therapeutic efficacy and to tune the formation of conjugates, a number of different PEGylation processes are now available to researchers for protein conjugation. Although the possibility of obtaining highly homogeneous conjugate mixtures, preferably formed by a single monoconjugate, from a chemical conjugation reaction continues to be limited, several enzymatic conjugation approaches have recently been investigated to address this need. PEGylation mediated by microbial transglutaminase and its many advantages and modifications are outlined in detail in the current work permitting interested readers to perform site-specific protein derivatization to glutamines or lysines.


Subject(s)
Bacterial Proteins/chemistry , Polyethylene Glycols/chemistry , Transglutaminases/chemistry , Bacterial Proteins/isolation & purification , Chromatography, High Pressure Liquid , Chromatography, Reverse-Phase , Enzyme Assays , Enzyme Stability , Enzymes, Immobilized/chemistry , Enzymes, Immobilized/isolation & purification , Kinetics , Transglutaminases/isolation & purification
18.
Bioconjug Chem ; 27(11): 2695-2706, 2016 Nov 16.
Article in English | MEDLINE | ID: mdl-27731976

ABSTRACT

Interferon α (IFN α) subtypes are important protein drugs that have been used to treat infectious diseases and cancers. Here, we studied the reactivity of IFN α-2b to microbial transglutaminase (TGase) with the aim of obtaining a site-specific conjugation of this protein drug. Interestingly, TGase allowed the production of two monoderivatized isomers of IFN with high yields. Characterization by mass spectrometry of the two conjugates indicated that they are exclusively modified at the level of Gln101 if the protein is reacted in the presence of an amino-containing ligand (i.e., dansylcadaverine) or at the level of Lys164 if a glutamine-containing molecule is used (i.e., carbobenzoxy-l-glutaminyl-glycine, ZQG). We explained the extraordinary specificity of the TGase-mediated reaction on the basis of the conformational features of IFN. Indeed, among the 10 Lys and 12 Gln residues of the protein, only Gln101 and Lys164 are located in highly flexible protein regions. The TGase-mediated derivatization of IFN was then applied to the production of IFN derivatives conjugated to a 20 kDa polyethylene glycol (PEG), using PEG-NH2 for Gln101 derivatization and PEG modified with ZQG for Lys164 derivatization. The two mono-PEGylated isomers of IFN were obtained in good yields, purified, and characterized in terms of protein conformation, antiviral activity, and pharmacokinetics. Both conjugates maintained a native-like secondary structure, as indicated by far-UV circular dichroism spectra. Importantly, they disclosed good in vitro antiviral activity retention (about only 1.6- to 1.8-fold lower than that of IFN) and half-lives longer (about 5-fold) than that of IFN after intravenous administration to rats. Overall, these results provide evidence that TGase can be used for the development of site-specific derivatives of IFN α-2b possessing interesting antiviral and pharmacokinetic properties.


Subject(s)
Glutamine/chemistry , Interferon-alpha/chemistry , Lysine/chemistry , Transglutaminases/metabolism , Amino Acid Sequence , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Binding Sites , Humans , Interferon alpha-2 , Interferon-alpha/pharmacokinetics , Interferon-alpha/pharmacology , Models, Molecular , Molecular Weight , Polyethylene Glycols/chemistry , Protein Structure, Secondary , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Substrate Specificity , Vesiculovirus/drug effects
19.
J Control Release ; 236: 79-89, 2016 08 28.
Article in English | MEDLINE | ID: mdl-27356018

ABSTRACT

While interferon alpha (IFNα) is used in several viral and cancer contexts, its efficacy against ovarian cancer (OC) is far from being incontrovertibly demonstrated and, more importantly, is hindered by heavy systemic side effects. To overcome these issues, here we propose a strategy that allows a targeted delivery of the cytokine, by conjugating IFNα2a with an aldehyde-modified form of hyaluronic acid (HA). The resulting HA-IFNα2a bioconjugate was biochemically and biologically characterized. The conjugation with HA did not substantially modified both the antiviral function and the anti-proliferative activity of the cytokine. Moreover, the induction of STAT1 phosphorylation and of a specific gene expression signature in different targets was retained. In vivo optical imaging biodistribution showed that the i.p.-injected HA-IFNα2a persisted into the peritoneal cavity longer than IFNα2a without being toxic for intraperitoneal organs, thus potentially enhancing the loco-regional therapeutic effect. Indeed, in OC xenograft mouse models bioconjugate significantly improved survival as compared to the free cytokine. Overall, HA-IFNα2a bioconjugate disclosed an improved anticancer efficacy, and can be envisaged as a promising loco-regional treatment for OC.


Subject(s)
Antineoplastic Agents/administration & dosage , Hyaluronic Acid/chemistry , Interferon-alpha/administration & dosage , Ovarian Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Antiviral Agents/administration & dosage , Antiviral Agents/chemistry , Cell Line, Tumor , Cell Survival , Female , Gene Expression , Humans , Interferon alpha-2 , Interferon-alpha/chemistry , Interferon-alpha/metabolism , Mice, Inbred BALB C , Phosphorylation , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , STAT1 Transcription Factor/metabolism , Tissue Distribution
20.
Macromol Biosci ; 16(1): 50-6, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26350165

ABSTRACT

The use of therapeutic proteins is often impaired by their short in vivo half-lives. PEGylation has been exploited to enhance protein stability and to prolong the pharmacokinetic. The biophysical characterization of two site-specific mono-PEGylated forms of human growth hormone (hGH)--chemically N-terminal PEGylated hGH (PEG-Nter-hGH) and enzymatically Gln141 PEGylated hGH (PEG-Gln141-hGH) via transglutaminase--is outlined here and their pharmacodynamics are compared. The thermal stability of PEG-Nter-hGH was increased with respect to that of hGH and PEG-Gln141-hGH. Pharmacodynamic studies in rats showed that a single injection of the conjugates had a better or comparable potency with respect to a daily hGH on a week schedule in terms of weight gain, femoral length, and tibial diaphysis width.


Subject(s)
Growth/drug effects , Human Growth Hormone/metabolism , Polyethylene Glycols/metabolism , Animals , Half-Life , Human Growth Hormone/chemistry , Human Growth Hormone/pharmacokinetics , Human Growth Hormone/pharmacology , Male , Polyethylene Glycols/chemistry , Protein Stability , Rats , Transglutaminases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL