Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Bioorg Chem ; 149: 107504, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38850783

ABSTRACT

The notable characteristics of recently emerged Antibody-Drug Conjugates (ADCs) encompass the targeting of Human Epidermal growth factor Receptor 2 (HER2) through monoclonal antibodies (mAbs) and a high ratio of drug to antibody (DAR). The achievements of Kadcyla® (T-DM1) and Enhertu® (T-Dxd) have demonstrated that HER2-targeting antibodies, such as trastuzumab, have shown to be competitive in terms of efficacy and price for development. Furthermore, with the arrival of T-Dxd and Trodelvy®, high-DAR (7-8) ADCs, which differ from the moderate DAR (3-4) ADCs that were formerly regarded as conventional, are being acknowledged for their worth. Following this trend of drug development, we endeavored to develop a high-DAR ADC using a straightforward approach involving the utilization of DM1, a highly potent substance, in combination with the widely recognized trastuzumab. To achieve a high DAR, DM1 was conjugated to reduced cysteine through the simple design and synthesis of various dimaleimide linkers with differing lengths. Using LC and MS analysis, we have demonstrated that our synthesis methodology is uncomplicated and efficacious, yielding trastuzumab-based ADCs that exhibit a remarkable degree of uniformity. These ADCs have been experimentally substantiated to exert an inhibitory effect on cancer cells in vitro, thus affirming their value as noteworthy additions to the realm of ADCs.


Subject(s)
Ado-Trastuzumab Emtansine , Immunoconjugates , Receptor, ErbB-2 , Trastuzumab , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Ado-Trastuzumab Emtansine/chemistry , Trastuzumab/chemistry , Trastuzumab/pharmacology , Molecular Structure , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Maleimides/chemistry , Maleimides/chemical synthesis , Dose-Response Relationship, Drug , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Structure-Activity Relationship , Maytansine/chemistry , Maytansine/pharmacology , Maytansine/chemical synthesis , Maytansine/analogs & derivatives , Cell Line, Tumor , Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/chemical synthesis , Antineoplastic Agents, Immunological/pharmacology
2.
Bioconjug Chem ; 32(8): 1525-1534, 2021 08 18.
Article in English | MEDLINE | ID: mdl-34105345

ABSTRACT

Antibody-drug conjugates (ADCs) have attracted great attention in recent years in the wake of an accelerated FDA approval rate and several large-scale acquisitions. To date, there are ten ADC drugs on the market and more than 70 in various stages of clinical trials. Yet, due to the complicated nature of ADC molecules, considerations need to cover many aspects for the success of ADCs, including target specificity, linker-payload stability, tumor permeability, and clearance rate. This topical review summarizes and discusses current methods used to increase stability and homogeneity of ADCs of cysteine conjugation. We believe that they will lead to improvement of efficacy and pharmacokinetics (PK) of ADC drugs.


Subject(s)
Cysteine/chemistry , Immunoconjugates/chemistry , Animals , Antineoplastic Agents, Immunological/chemical synthesis , Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/pharmacokinetics , Chemistry Techniques, Synthetic/methods , Cysteine/chemical synthesis , Cysteine/pharmacokinetics , Humans , Immunoconjugates/pharmacokinetics , Neoplasms/drug therapy
3.
Cancer Med ; 10(6): 2125-2136, 2021 03.
Article in English | MEDLINE | ID: mdl-33629544

ABSTRACT

BACKGROUND: CD47 is a widely expressed transmembrane protein located on the surface of somatic cells. It mediates a variety of cellular processes including apoptosis, proliferation, adhesion, and migration. An important role for CD47 is the transmission of a "Don't eat me" signal by interacting with SIRPα on the macrophage surface membrane, thereby preventing the phagocytosis of normal cells. However, cancer cells can take advantage of this autogenous signal to protect themselves from phagocytosis, thus enabling immune escape. Blocking the interaction between CD47 and SIRPα has proven to be effective in removing cancer cells. The treatment of various cancers with CD47 monoclonal antibodies has also been validated. METHODS: We designed and synthesized a peptide (RS17), which can specifically bind to CD47 and block CD47-SIRPα signaling. The affinity of RS17 for CD47-expressing tumor cells was determined, while the inhibition of CD47-SIRPα signaling was evaluated in vitro and in vivo. RESULTS: The results indicated that RS17 significantly promotes the phagocytosis of tumor cells by macrophages and had a similar therapeutic effect compared with a positive control (CD47 monoclonal antibodies). In addition, a cancer xenograft mouse model was established using CD47-expressing HepG2 cells to evaluate the effect of RS17 on tumor growth in vivo. Using ex vivo and in vivo mouse models, RS17 demonstrated a high inhibitory effect on tumor growth. CONCLUSIONS: Based on our results, RS17 may represent a novel therapeutic peptide for cancer therapy.


Subject(s)
Antigens, Differentiation/metabolism , Antineoplastic Agents, Immunological/metabolism , CD47 Antigen/metabolism , Macrophages/metabolism , Peptides/metabolism , Phagocytosis , Receptors, Immunologic/metabolism , Amino Acids/metabolism , Animals , Antibodies, Monoclonal , Antineoplastic Agents, Immunological/chemical synthesis , Antineoplastic Agents, Immunological/therapeutic use , CD47 Antigen/antagonists & inhibitors , CD47 Antigen/chemistry , Carcinoma, Squamous Cell/metabolism , Cell Culture Techniques , Cell Line, Tumor , Female , Flow Cytometry , Hep G2 Cells/metabolism , Humans , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Docking Simulation , Molecular Targeted Therapy/methods , Peptides/chemical synthesis , Peptides/therapeutic use , Phagocytosis/immunology , Skin Neoplasms/metabolism , Software , Tumor Escape , Xenograft Model Antitumor Assays
4.
ChemMedChem ; 16(3): 568-577, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33085193

ABSTRACT

The interaction between programmed cell death-1 (PD-1) and its ligand PD-L1 activates a coinhibitory signal that blocks T-cell activation, promoting the immune escape process in the tumor microenvironment. Development of monoclonal antibodies targeting and inhibiting PD-1/PD-L1 interaction as anticancer immunotherapies has proved successful in multiple clinical settings and for various types of cancer. Notwithstanding, limitations exist with the use of these biologics, including drug resistance and narrow therapeutic response rate in a majority of patients, that demand for the design of more efficacious small molecule-based immunotherapies. Alteration of pH in the tumor microenvironment is a key factor that is involved in promoting drug resistance, tumor survival and progression. In this study, we have investigated the effect of pH shifts on binding properties of distinct classes of PD-L1 inhibitors, including macrocyclic peptide and small molecules. Results expand structure-activity relationships of PD-L1 inhibitors, providing insights into structural features and physicochemical properties that are useful for the design of ligands that may escape a drug resistance mechanism associated to variable pH conditions of tumor microenvironment.


Subject(s)
Antibodies, Monoclonal/metabolism , Antineoplastic Agents, Immunological/pharmacology , B7-H1 Antigen/antagonists & inhibitors , Immune Checkpoint Inhibitors/pharmacology , Neoplasms/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Antibodies, Monoclonal/chemistry , Antineoplastic Agents, Immunological/chemical synthesis , Antineoplastic Agents, Immunological/chemistry , B7-H1 Antigen/metabolism , Dose-Response Relationship, Drug , Humans , Hydrogen-Ion Concentration , Immune Checkpoint Inhibitors/chemical synthesis , Immune Checkpoint Inhibitors/chemistry , Immunotherapy , Models, Molecular , Molecular Structure , Neoplasms/metabolism , Programmed Cell Death 1 Receptor/metabolism , Structure-Activity Relationship
5.
J Am Chem Soc ; 142(32): 13657-13661, 2020 08 12.
Article in English | MEDLINE | ID: mdl-32706963

ABSTRACT

This communication describes the design, synthesis, and biological activity of a megamolecule mimic of an anti-HER2 antibody. The antibody mimic was prepared by linking two Fabs from the therapeutic antibody trastuzumab, which are fused through the heavy chain variable domain to either cutinase or SnapTag, with a linker terminated in an irreversible inhibitor for each enzyme. This mimic binds HER2 with comparable avidity to trastuzumab, has similar activity in a cell-based assay, and can arrest tumor growth in a mouse xenograft BT474 tumor model. A panel of 16 bivalent anti-HER2 antibodies were prepared wherein each varied in the orientation of the fusion domain on the Fabs. The analogs displayed a range of cytotoxic activity, and surprisingly, the most active mimic binds to cells with a 10-fold lower avidity than the least active variant suggesting that structure plays a large role in their efficacy. This work suggests that the megamolecule approach can be used to prepare antibody mimics having a broad structural diversity.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Drug Design , Receptor, ErbB-2/antagonists & inhibitors , Trastuzumab/pharmacology , Animals , Antineoplastic Agents, Immunological/chemical synthesis , Antineoplastic Agents, Immunological/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Female , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mice , Mice, SCID , Molecular Structure , Trastuzumab/chemistry
6.
Eur J Med Chem ; 169: 29-41, 2019 May 01.
Article in English | MEDLINE | ID: mdl-30852385

ABSTRACT

Increasing evidences suggested that cisplatin can be involved in a tumor-specific immune response as an immunomodulator to improve antitumor immunity, but the designation and development are limited. Here, we report a series of novel Pt(IV) complexes derived from the conjugation of platinum(II) anticancer agents with an immune checkpoint TDO inhibitor. These complexes not only showed significant cytotoxic effects on the tested cancer cell lines, but also could enhance antitumor immune response. Particularly, complex T2, the mono-conjuagte of oxoplatin and (E)-4-oxo-4-(3-(2-(pyridin-3-yl)vinyl)-1H-indol-1-yl)butanoic acid, displayed 35-fold more potency than cisplatin against TDO-overexpressed HepG-2 cancer cells. Further study indicated that T2 could inhibit TDO via releasing a derivative of a TDO inhibitor and block kynurenine production, resulting in T-cell activation and proliferation in vitro. In vivo tests proved that T2 as a potent immunomodulator could highly promote the antitumor activity of cisplatin and effectively suppress the expression of TDO. This immunochemotherapeutic strategy can be promisingly applied to treat with TDO-overexpressed cancers.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Enzyme Inhibitors/pharmacology , Immunologic Factors/pharmacology , Organoplatinum Compounds/pharmacology , Tryptophan Oxygenase/antagonists & inhibitors , Animals , Antineoplastic Agents, Immunological/chemical synthesis , Antineoplastic Agents, Immunological/chemistry , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Immunologic Factors/chemistry , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Organoplatinum Compounds/chemical synthesis , Organoplatinum Compounds/chemistry , Structure-Activity Relationship , Tryptophan Oxygenase/metabolism
7.
Sci Rep ; 9(1): 2443, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30792442

ABSTRACT

Antibody-drug conjugates (ADCs) are promising therapies for haematological cancers. Historically, their therapeutic benefit is due to ADC targeting of lineage-restricted antigens. The C-X-C motif chemokine receptor 4 (CXCR4) is attractive for targeted therapy of haematological cancers, given its expression in multiple tumour types and role in cancer "homing" to bone marrow. However, CXCR4 is also expressed in haematopoietic cells and other normal tissues, raising safety challenges to the development of anti-CXCR4 ADCs for cancer treatment. Here, we designed the first anti-CXCR4 ADC with favourable therapeutic index, effective in xenografts of haematopoietic cancers resistant to standard of care and anti-CXCR4 antibodies. We screened multiple ADC configurations, by varying type of linker-payload, drug-to-antibody ratio (DAR), affinity and Fc format. The optimal ADC bears a non-cleavable linker, auristatin as payload at DAR = 4 and a low affinity antibody with effector-reduced Fc. Contrary to other drugs targeting CXCR4, anti-CXCR4 ADCs effectively eliminated cancer cells as monotherapy, while minimizing leucocytosis. The optimal ADC selectively eliminated CXCR4+ cancer cells in solid tumours, but showed limited toxicity to normal CXCR4+ tissues, sparing haematopoietic stem cells and progenitors. Our work provides proof-of-concept that through empirical ADC design, it is possible to target proteins with broad normal tissue expression.


Subject(s)
Antineoplastic Agents, Immunological , Drug Design , Immunoconjugates , Receptors, CXCR4/immunology , Animals , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/chemical synthesis , Antineoplastic Agents, Immunological/chemistry , CHO Cells , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cells, Cultured , Cricetinae , Cricetulus , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Deoxycytidine/analogs & derivatives , Female , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/adverse effects , Immunoconjugates/chemistry , Immunoglobulin Fab Fragments/administration & dosage , Immunoglobulin Fab Fragments/adverse effects , Immunoglobulin Fab Fragments/chemistry , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, SCID , Mice, Transgenic , Models, Molecular , Protein Structure, Tertiary , Receptors, CXCR4/antagonists & inhibitors , Treatment Outcome , Xenograft Model Antitumor Assays , Gemcitabine
9.
Med Sci (Paris) ; 35(12): 1202-1203, 2019 Dec.
Article in French | MEDLINE | ID: mdl-31903940
10.
J Pharm Sci ; 107(7): 1858-1869, 2018 07.
Article in English | MEDLINE | ID: mdl-29626535

ABSTRACT

The commercially available antibody-drug conjugate (ADC) product, Kadcyla® is synthesized using a 2-step reaction, wherein the linker is conjugated to native lysines on the mAb in step 1, followed by drug conjugation to the linker-modified antibody in step 2. In our study, we synthesized a lysine-conjugated ADC (Syn-ADC) on the same trastuzumab scaffold as Kadcyla® using a 1-step reaction. Mass spectrometry of both products revealed a subpopulation of Kadcyla® containing free linkers conjugated to the mAb, but not conjugated to the drug, which were absent in the 1-step reaction ADC product. Differential scanning calorimetry thermograms showed that the drug and linker conjugation significantly reduced the thermal stability and energies of activation for the denaturation of the CH2 domain of the ADCs. The heating induced aggregation events started as early as ∼57°C and ∼45°C for Kadcyla® and Syn-ADC, respectively, compared with 71°C for Herceptin®. The colloidal stability measurements clearly showed that the hydrophobic drug payload on ADCs significantly reduced the repulsive interprotein interactions when compared to the unconjugated antibody under formulation buffer conditions (pH 6.0). Attaching hydrophobic drug and linker moieties onto the antibody lowered the thermal and colloidal stabilities and increased the aggregation propensity of the ADCs.


Subject(s)
Antineoplastic Agents, Immunological/chemistry , Immunoconjugates/chemistry , Lysine/chemistry , Maytansine/analogs & derivatives , Protein Aggregates , Trastuzumab/chemistry , Ado-Trastuzumab Emtansine , Antineoplastic Agents, Immunological/chemical synthesis , Calorimetry, Differential Scanning , Chromatography, Liquid , Circular Dichroism , Colloids/chemical synthesis , Colloids/chemistry , Heating , Lysine/chemical synthesis , Mass Spectrometry , Maytansine/chemical synthesis , Maytansine/chemistry , Protein Stability , Spectrum Analysis, Raman
11.
Org Biomol Chem ; 16(11): 1882-1889, 2018 03 14.
Article in English | MEDLINE | ID: mdl-29473076

ABSTRACT

Herein we describe the synthesis and evaluation of four novel HER2-targeting, cathepsin B-sensitive antibody-drug conjugates bearing a monomethylauristatin E (MMAE) cytotoxic payload, constructed via the conjugation of cleavable linkers to trastuzumab using a site-specific bioconjugation methodology. These linkers vary by both cleavable trigger motif and hydrophilicity, containing one of two cathepsin B sensitive dipeptides (Val-Cit and Val-Ala), and engendered with either hydrophilic or hydrophobic character via application of a PEG12 spacer. Through evaluation of physical properties, in vitro cytotoxicity, and receptor affinity of the resulting antibody-drug conjugates (ADCs), we have demonstrated that while both dipeptide triggers are effective, the increased hydrophobicity of the Val-Ala pair limits its utility within this type of linker. In addition, while PEGylation augments linker hydrophilicity, this change does not translate to more favourable ADC hydrophilicity or potency. While all described structures demonstrated excellent and similar in vitro cytotoxicity, the ADC with the ValCitPABMMAE linker shows the most promising combination of in vitro potency, structural homogeneity, and hydrophilicity, warranting further evaluation into its therapeutic potential.


Subject(s)
Antineoplastic Agents, Immunological/chemistry , Cathepsin B/metabolism , Immunoconjugates/chemistry , Oligopeptides/chemistry , Receptor, ErbB-2/metabolism , Trastuzumab/chemistry , Antineoplastic Agents, Immunological/chemical synthesis , Antineoplastic Agents, Immunological/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Chemistry Techniques, Synthetic/methods , Drug Delivery Systems , Humans , Hydrophobic and Hydrophilic Interactions , Immunoconjugates/pharmacology , Neoplasms/drug therapy , Oligopeptides/chemical synthesis , Oligopeptides/pharmacology , Trastuzumab/pharmacology
12.
Blood ; 131(7): 746-758, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29284597

ABSTRACT

B-cell maturation antigen (BCMA) is a promising therapeutic target for multiple myeloma (MM), but expression is variable, and early reports of BCMA targeting chimeric antigen receptors (CARs) suggest antigen downregulation at relapse. Dual-antigen targeting increases targetable tumor antigens and reduces the risk of antigen-negative disease escape. "A proliferation-inducing ligand" (APRIL) is a natural high-affinity ligand for BCMA and transmembrane activator and calcium-modulator and cyclophilin ligand (TACI). We quantified surface tumor expression of BCMA and TACI on primary MM cells (n = 50). All cases tested expressed BCMA, and 39 (78%) of them also expressed TACI. We engineered a third-generation APRIL-based CAR (ACAR), which killed targets expressing either BCMA or TACI (P < .01 and P < .05, respectively, cf. control, effector-to-target [E:T] ratio 16:1). We confirmed cytolysis at antigen levels similar to those on primary MM, at low E:T ratios (56.2% ± 3.9% killing of MM.1s at 48 h, E:T ratio 1:32; P < .01) and of primary MM cells (72.9% ± 12.2% killing at 3 days, E:T ratio 1:1; P < .05, n = 5). Demonstrating tumor control in the absence of BCMA, we maintained cytolysis of primary tumor expressing both BCMA and TACI in the presence of a BCMA-targeting antibody. Furthermore, using an intramedullary myeloma model, ACAR T cells caused regression of an established tumor within 2 days. Finally, in an in vivo model of tumor escape, there was complete ACAR-mediated tumor clearance of BCMA+TACI- and BCMA-TACI+ cells, and a single-chain variable fragment CAR targeting BCMA alone resulted in outgrowth of a BCMA-negative tumor. These results support the clinical potential of this approach.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , B-Cell Maturation Antigen/metabolism , Multiple Myeloma/drug therapy , Receptors, Chimeric Antigen/therapeutic use , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , Animals , Antineoplastic Agents, Immunological/chemical synthesis , Antineoplastic Agents, Immunological/chemistry , Cell Line, Tumor , Cytotoxicity, Immunologic , Humans , Ligands , Mice , Molecular Targeted Therapy , Receptors, Chimeric Antigen/chemical synthesis , Receptors, Chimeric Antigen/chemistry , Transmembrane Activator and CAML Interactor Protein/chemistry , Tumor Necrosis Factor Ligand Superfamily Member 13/chemistry
13.
Chembiochem ; 19(5): 459-469, 2018 03 02.
Article in English | MEDLINE | ID: mdl-29230922

ABSTRACT

Multispecific and multivalent antibodies are seen as promising cancer therapeutics, and numerous antibody fragments and derivatives have been developed to exploit avidity effects that result in increased selectivity. Most of these multispecific and multivalent antibody strategies make use of recombinant expression of antigen-binding modules. In contrast, chemical synthesis and chemoselective ligations can be used to generate a variety of molecules with different numbers and combinations of binding moieties in a modular and homogeneous fashion. In this study we synthesized a series of targeted immune system engagers (ISErs) by using solid-phase peptide synthesis and chemoselective ligations. To explore avidity effects, we constructed molecules bearing different numbers and combinations of two "binder" peptides that target ephrin A2 and integrin α3 receptors and an "effector" peptide that binds to formyl peptide receptors and stimulates an immune response. We investigated various strategies for generating multivalent and multispecific targeted innate immune stimulators and studied their activities in terms of binding to cancer cells and stimulation of immune cells. This study gives insights into the influence that multivalency and receptor density have on avidity effects and is useful for the design of potential anticancer therapeutics.


Subject(s)
Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacology , Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/pharmacology , Peptides/chemistry , Peptides/pharmacology , Adjuvants, Immunologic/chemical synthesis , Antineoplastic Agents, Immunological/chemical synthesis , Cell Line, Tumor , Humans , Immunity, Innate/drug effects , Neoplasms/drug therapy , Neoplasms/immunology , Peptides/chemical synthesis , Solid-Phase Synthesis Techniques
14.
Rev. eletrônica enferm ; 20: 1-17, 2018. ilus
Article in English, Portuguese | BDENF - Nursing, LILACS | ID: biblio-1118920

ABSTRACT

Este estudo objetivou identificar as evidências científicas disponíveis acerca das interações medicamentosas entre antineoplásicos decorrentes da ordem de infusão para, então, descrever a melhor sequência de aplicação e discutir sua aplicabilidade na Sistematização da Assistência de Enfermagem (SAE). Revisão integrativa da literatura operacionalizada em 2018 nas bases MEDLINE, LILACS, CINAHL e BVS, a partir dos termos Neoplasms, Drug Therapy, Drug Interactions,Chemotherapye Sequence Administration. Foram analisados 57 estudos que, em conjunto, estudaram 40 combinações de antineoplásicos, apontando as interações farmacocinéticas e farmacodinâmicas decorrentes da ordem de infusão, as quais subsidiaram a construção de um quadro determinando o melhor sequenciamento para cada uma dessas combinações. Um fluxograma também foi produzido para, junto com o quadro, apoiar a SAE na perspectiva da prática de enfermagem oncológica baseada em evidência. Selecionar as sequências de infusão de antineoplásicos combinados representa nova estratégia conceitual para enfermeiros que administram protocolos de poliquimioterapia.


The objective of this study was to find scientific evidence of drug interactions between antineoplastic drugs that result from the administration sequence and then describe the best sequence and discuss its applicability to nursing care systematization (NCS). An integrative review of the literature was carried out in 2018 in the MEDLINE, LILACS, CINAHL, and BVS databases, with the terms neoplasms, drug Therapy, drug Interactions, chemotherapy, and sequence of administration. Fifty-seven studies were analyzed, which, as a set, studied 40 combinations of antineoplastic drugs and found pharmacokinetic and pharmacodynamic interactions resulting from the sequence of administration, which supported the construction of a chart that indicates the bestsequence for each of those combinations. Along with the chart, a flowchart was also made to support NCS in the context of evidence-based oncology nursing practice. Selecting the sequences of antineoplastic drug administration is a new conceptual strategy designed for nurses who carry out multidrug therapy.


Subject(s)
Drug Interactions , Drug Therapy, Combination , Antineoplastic Agents, Immunological/chemical synthesis , Oncology Nursing , Neoplasms/drug therapy
15.
Curr Top Med Chem ; 17(32): 3444-3462, 2017.
Article in English | MEDLINE | ID: mdl-29357803

ABSTRACT

The recent approval of trastuzumab emtansine (Kadcyla®) and brentuximab vedotin (Adcetris ®) has spurred tremendous investment in new approaches for the targeted delivery of pharmaceutical agents. Targeted delivery approaches, such as Antibody Drug Conjugates (ADCs), typically rely on an endogenous or exogenous "trigger" that results in the release of the pharmacologically active agent at the intended site of action. Lysosomal and intracellular triggers include proteolytic cleavage, glycolytic cleavage, phosphatase cleavage, hydrolytic cleavage, and reductive cleavage. Recent work has also illustrated that exogenous triggers and extracellular enzymes can be harnessed to result in linker cleavage at the site of action. As these linker technologies have grown, so also has our understanding of the biophysical parameters that drive exposure and stability. The growth in targeted delivery approaches has also driven advancement in bioanalytical strategies for assessing the distribution, processing, and metabolism of these agents. This review provides a systematic overview of each of these areas, particularly focusing on recent advancements in the field that has the potential to expand the scope of therapeutic areas that ADCs and other targeted delivery approaches can be designed to address.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Biopharmaceutics , Cross-Linking Reagents/metabolism , Drug Delivery Systems , Drug Design , Immunoconjugates/therapeutic use , Maytansine/analogs & derivatives , Neoplasms/drug therapy , Trastuzumab/therapeutic use , Ado-Trastuzumab Emtansine , Antineoplastic Agents, Immunological/chemical synthesis , Antineoplastic Agents, Immunological/chemistry , Brentuximab Vedotin , Humans , Immunoconjugates/chemistry , Maytansine/chemistry , Maytansine/therapeutic use , Trastuzumab/chemistry
16.
J Labelled Comp Radiopharm ; 60(1): 12-19, 2017 01.
Article in English | MEDLINE | ID: mdl-27813128

ABSTRACT

Trastuzumab that targets the human epidermal growth factor receptor type 2 (HER2) is known to benefit patients with HER2+ metastatic breast cancer. The objective was to explore the potential of 131 I-trastuzumab for treatment of breast cancers. Radioiodination of trastuzumab was carried out by chloramine-T method, purified by using PD-10 column, and characterized by size exclusion high-performance liquid chromatography on a gel column. In vitro studies were carried out in HER2+ cells to determine the specificity of the radioimmunoconjugate. Uptake and retention of 131 I-trastuzumab were determined by biodistribution studies in tumor-bearing non-obese diabetic/severe combined immunodeficiency and normal severe combined immunodeficiency mice. The radiochemical purity (RCP) of 131 I-trastuzumab was 98 ± 0.4% with retention time of 17 minutes by high-performance liquid chromatography. In vitro stability studies exhibited RCP of more than 90% in serum at 37°C after 120 hours of radioiodination. In vitro cell binding with 131 I-trastuzumab in HER2+ cells showed binding of 28% to 35% which was inhibited significantly, with unlabeled trastuzumab confirming its specificity. Kd value of 131 I-trastuzumab was 0.5 nM, while its immunoreactivity was more than 80%. Uptake of more than 12% and retention were observed in the tumors up to 120 hours p.i. 131 I-trastuzumab prepared in-house-exhibited RCP of more than 98%, excellent immunoreactivity, affinity to HER2+ cell lines and good tumor uptake thereby indicating its potential for further evaluation in HER2+ breast cancers.


Subject(s)
Antineoplastic Agents, Immunological/chemical synthesis , Iodine Radioisotopes/chemistry , Radiopharmaceuticals/chemical synthesis , Trastuzumab/chemistry , Animals , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/pharmacokinetics , Cell Line, Tumor , Female , Humans , Mammary Neoplasms, Experimental/drug therapy , Mice , Mice, Inbred NOD , Mice, SCID , Protein Binding , Radiopharmaceuticals/adverse effects , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Receptor, ErbB-2/metabolism , Tissue Distribution
17.
Photochem Photobiol Sci ; 15(10): 1227-1238, 2016 10 05.
Article in English | MEDLINE | ID: mdl-27501936

ABSTRACT

In many cancers early intervention involves surgical resection of small localised tumour masses. Inadequate resection leads to recurrence whereas overzealous treatment can lead to organ damage. This work describes production of a HER2 targeting antibody Fab fragment dual conjugated to achieve both real time near-infrared fluorescent imaging and photodynamic therapy. The use of fluorescence emission from a NIR-dye could be used to guide resection of tumour bulk, for example during endoscopic diagnosis for oesophago-gastric adenocarcinoma, this would then be followed by activation of the photodynamic therapeutic agent to destroy untreated localised areas of cancer infiltration and tumour infiltrated lymph nodes. This theranostic agent was prepared from the Fab fragment of trastuzumab initially by functional disulfide re-bridging and site-specific click reaction of a NIR-dye. This was followed by further reaction with a novel pre-activated form of the photosensitiser chlorin e6 with the exposed fragments' lysine residues. Specific binding of the theranostic agent was observed in vitro with a HER2 positive cell line and cellular near-infrared fluorescence was observed with flow cytometry. Specific photo-activity of the conjugates when exposed to laser light was observed with HER2 positive but not HER2 negative cell lines in vitro, this selectivity was not seen with the unconjugated drug. This theranostic agent demonstrates that two different photo-active functions can be coupled to the same antibody fragment with little interference to their independent activities.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Neoplasms/drug therapy , Photochemotherapy , Photosensitizing Agents/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Theranostic Nanomedicine , Trastuzumab/pharmacology , Antineoplastic Agents, Immunological/chemical synthesis , Antineoplastic Agents, Immunological/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Neoplasms/metabolism , Neoplasms/pathology , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/chemistry , Receptor, ErbB-2/metabolism , Structure-Activity Relationship , Trastuzumab/chemistry , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...