11 research outputs found

    Optimisation of culture conditions for a producer clone coexpressing arylsulfatase B and a formylglycine-generating enzyme in order to increase the yield of arylsulfatase B

    Get PDF
    Maroteaux—Lamy syndrome (mucopolysaccharidosis type VI) is an orphan genetic disease caused by mutations in the arylsulfatase B gene (ARSB), which encodes the lysosomal enzyme arylsulfatase B (ASB). The relevance of the study lies in the need of a Russian recombinant ASB product for patients with the disease in the Russian Federation. Previously, the authors have developed producer lines coexpressing the target ASB enzyme with an auxiliary formylglycine-generating enzyme (FGE), based on Chinese hamster ovary (CHO) cells. Further development of the recombinant ASB preparation places priority on increasing the enzyme yield. The aim of this study was to increase the productivity of producer clones by optimising the culture process and adding calcium chloride and copper sulfate to the culture medium. Materials and methods: a suspension-adapted CHO cell line was used. Monoclonal cell lines were developed using Cell Metric and ClonePix FL systems. The concentration of ASB in the culture liquid was determined using the enzyme-linked immunosorbent assay (ELISA). The authors analysed batch culture and/or fed-batch culture in media supplemented with various concentrations of copper sulfate and calcium chloride. Results: the combined addition of copper sulfate and calcium chloride at concentrations of 300 μM during batch culture of producer clones coexpressing ASB and FGE increases viability and specific productivity of the cells up to 4.58±1.62 pg/ (cell×day). The cultivation of the lead producer clone coexpressing ASB and FGE under fed-batch conditions for 12 days and the addition of copper sulfate to the growth medium at the concentration of 300 μM allow for increasing the yield of the active lysosomal enzyme, arylsulfatase B, to 420 mg/L. Conclusions: the cultivation of producer clones coexpressing ASB and FGE under fed-batch conditions with copper sulfate added to the medium significantly improves cell line growth properties and the ASB yield. This approach to the selection of culture conditions for producer cell lines can be applied to other enzymes of the sulfatase family

    Интернализация рекомбинантной имиглюцеразы в перитонеальные макрофаги мыши и фибробласты мыши линии L929

    Get PDF
    Enzyme replacement therapy (ERT) is one of the most efficient treatments for lysosomal storage diseases. Type 1 Gaucher disease is caused by β-glucocerebrosidase enzyme deficiency, which may be compensated for by intravenous infusions of imiglucerase—a recombinant enzyme. Imiglucerase targets macrophages and enters these cells via interaction with mannose receptors on the cell membrane. Characterisation of internalization of enzymes by target cells is important in the context of the development of new medicines and production of existing ERT medicines. The peritoneal and alveolar macrophages, as well as macrophages of the spleen of small laboratory animals (rats and mice) are widely used in such studies. However, isolation of cells from animal sources raises ethical issues, and therefore continuous mammalian cell lines may offer an attractive alternative. The aim of the study: to conduct comparative studies on the internalization of recombinant imiglucerase into mouse peritoneal macrophages and L929 mouse fibroblasts. Materials and methods: CerezymeR batches 7HV0913, C6214H05, 7HV0888 (Genzyme Ltd., UK); Glurazim batches 020416, 011117, 021117 (LLC “IBC “Generium”, Russia). We used peritoneal macrophages obtained from BALB/c mice and L929 mouse fibroblasts. The cells were cultured in DMEM/F12 complete growth medium with 10% fetal bovine serum. The activity of imiglucerase internalized into the cells was evaluated spectrophotometrically by hydrolysis of the artificial substrate—4-methylumbelliferyl-β-Dglucopyranoside. Results: the study compared internalization of recombinant imiglucerase (the active ingredient of CerezymeR and Glurazim) by mouse peritoneal macrophages and L929 mouse fibroblasts. It was demonstrated that the medicines activity in the lysates of peritoneal macrophages is comparable with that in the lysates of L929 mouse fibroblasts. Regardless of the model system, the activity of Glurazim stayed within the acceptable range (80–125%) established for biosimilar products. Conclusions: the experiments proved that L929 mouse fibroblasts could be recommended for assessment of internalization of recombinant imiglucerase.Фермент-заместительная терапия (ФЗТ) является одной из самых действенных при лечении болезней лизосомального накопления. Болезнь Гоше первого типа характеризуется недостатком нативного фермента β-глюкоцереброзидазы, который возмещают внутривенными инфузиями рекомбинантного фермента (имиглюцераза). Клетками-мишенями имиглюцеразы являются макрофаги, в которые фермент проникает посредством взаимодействия с рецепторами маннозы на клеточной мембране. Оценка интернализации ферментов клетками-мишенями представляет интерес при разработке новых и воспроизведении существующих препаратов для ФЗТ. Для этих исследований широко применяются перитонеальные и альвеолярные макрофаги, макрофаги селезенки мелких лабораторных животных (крыс и мышей). Однако получение таких клеток затрагивает этические вопросы использования лабораторных животных. Альтернативой являются перевиваемые клеточные линии млекопитающих. Цель работы: провести сравнительные исследования интернализации рекомбинантной имиглюцеразы в перитонеальные макрофаги мыши и фибробласты мыши линии L929. Материалы и методы: Церезим®, серии 7HV0913, C6214H05, 7HV0888 (Джензайм Лтд., Великобритания); Глуразим, серии 020416, 011117, 021117 (ООО «МБЦ «Генериум», Россия). В работе использовали перитонеальные макрофаги, полученные от мышей линии BALB/c, и фибробласты мыши линии L929. Клетки культивировали в полной ростовой среде ДМЕМ/Ф12 c добавлением 10% сыворотки плода крупного рогатого скота. Активность имиглюцеразы, проникшей в клетки, оценивали спектрофотометрически по гидролизу искусственного субстрата 4-метилумбеллиферил-β-D-глюкопиранозида. Результаты: представлены данные сравнительной оценки интернализации рекомбинантной имиглюцеразы, действующего вещества препаратов Церезим® и Глуразим, перитонеальными макрофагами мыши и клетками фибробластов мыши линии L929. Показано, что активность препаратов в лизатах перитонеальных макрофагов сопоставима с их активностью в лизатах клеток фибробластов мыши линии L929, при этом активность разработанного препарата Глуразим независимо от типа клеток, была в границах допустимого диапазона (80–125%), установленного для биоподобных препаратов. Выводы: экспериментально доказано, что фибробласты мыши линии L929 могут быть рекомендованы для оценки интернализации рекомбинантной имиглюцеразы

    Новый высокоэффективный ИК-фотосенсибилизатор для фотодинамической терапии рака

    Get PDF
    The present article is devoted to the synthesis and investigation of biological and physico-chemical properties of the bacteriopurpurinimide derivative that was used as a photosensitizer. These substances have an ability to accumulate selectively in the malignant tumours. Also this class of photosensitizers is of particular interest because of their strong light absorbtion spectrum in red and NIR-region in the “window of tissue transparency”. This property affords to treat deep-seated tumours at the depth of 2 cm. In this study it was implemented the method of photodynamic therapy of cancer. The photosensitizer for this study was prepared as the nanostructured water-soluble micellar dispersion of O-propyloxime-N-propoxybacteriopurpurinimide methyl ester. The photodynamic therapy using this substance provided the highly effective photoinduced antitumour in vitro and in vivo activity, 100% regression of the tumour and 90% cure of animals.Осуществлен синтез и изучены физико-химические и биологические свойства производного бактерио-хлорофилла а - метилового эфира О-пропилоксима N-пропоксибактериопурпуринимида, использован-ного в качестве фотосенсибилизатора в виде наноструктурированной водной дисперсии. В работе реализован метод фотодинамической терапии рака, который включает системное введение данного фотосенсибилизатора в дозах от 1.0 до 10.0 мг/кг и облучение патологического участка оптическим излучением через 0.25-8 ч после введения препарата в спектральном диапазоне 789-831 нм при плотности энергии от 45 до 360 Дж/см2. Показано, что фотодинамическая терапия с использованием данного препарата обеспечивает высокую фотоиндуцированную противоопухолевую активность в системе in vitro и in vivo, 100%-ное торможение роста опухоли и 90%-ную излеченность животных за счет селективного накопления в опухоли и быстрого выведения из организма

    Оптимизация условий культивирования клона-продуцента, коэкспрессирующего арилсульфатазу B и формилглицин-генерирующий фермент, с целью повышения выхода фермента арилсульфатазы B

    Get PDF
    Maroteaux—Lamy syndrome (mucopolysaccharidosis type VI) is an orphan genetic disease caused by mutations in the arylsulfatase B gene (ARSB), which encodes the lysosomal enzyme arylsulfatase B (ASB). The relevance of the study lies in the need of a Russian recombinant ASB product for patients with the disease in the Russian Federation. Previously, the authors have developed producer lines coexpressing the target ASB enzyme with an auxiliary formylglycine-generating enzyme (FGE), based on Chinese hamster ovary (CHO) cells. Further development of the recombinant ASB preparation places priority on increasing the enzyme yield. The aim of this study was to increase the productivity of producer clones by optimising the culture process and adding calcium chloride and copper sulfate to the culture medium. Materials and methods: a suspension-adapted CHO cell line was used. Monoclonal cell lines were developed using Cell Metric and ClonePix FL systems. The concentration of ASB in the culture liquid was determined using the enzyme-linked immunosorbent assay (ELISA). The authors analysed batch culture and/or fed-batch culture in media supplemented with various concentrations of copper sulfate and calcium chloride. Results: the combined addition of copper sulfate and calcium chloride at concentrations of 300 μM during batch culture of producer clones coexpressing ASB and FGE increases viability and specific productivity of the cells up to 4.58±1.62 pg/ (cell×day). The cultivation of the lead producer clone coexpressing ASB and FGE under fed-batch conditions for 12 days and the addition of copper sulfate to the growth medium at the concentration of 300 μM allow for increasing the yield of the active lysosomal enzyme, arylsulfatase B, to 420 mg/L. Conclusions: the cultivation of producer clones coexpressing ASB and FGE under fed-batch conditions with copper sulfate added to the medium significantly improves cell line growth properties and the ASB yield. This approach to the selection of culture conditions for producer cell lines can be applied to other enzymes of the sulfatase family.Синдром Марото—Лами (мукополисахаридоз VI типа) — орфанное генетическое заболевание, вызванное мутациями гена ARSB, который кодирует лизосомальный фермент арилсульфатазу В. Актуальность исследования заключается в необходимости разработки отечественного препарата рекомбинантной арилсульфатазы В для лечения пациентов с данным заболеванием в Российской Федерации. Ранее были получены клеточные линии-продуценты, коэкспрессирующие целевой фермент арилсульфатазу В и вспомогательный формилглицин-генерирующий фермент на основе клеточной линии СНО. Однако для дальнейшей разработки препарата рекомбинантной арилсульфатазы В представляется важным повышение выхода фермента. Цель работы: увеличение продуктивности клонов-продуцентов за счет оптимизации процесса культивирования и добавления в культуральную среду хлорида кальция и сульфата меди. Материалы и методы: использовали суспензионную клеточную линию СНО. Моноклональные клеточные линии получали с использованием систем Cell Metric и Clone Pix FL. Концентрацию арилсульфатазы В в культуральной жидкости определяли методом иммуноферментного анализа. Использовали периодическое культивирование (batch culture) и/или периодическое культивирование с подпиткой (fedbatch culture) в среде с добавлением различных концентраций сульфата меди и хлорида кальция. Результаты: продемонстрировано, что одновременное добавление сульфата меди и хлорида кальция в концентрации 300 мкM при периодическом культивировании клонов-продуцентов, коэкспрессирующих арилсульфатазу В и формилглицин-генерирующий фермент, увеличивает жизнеспособность культур и повышает удельную продуктивность клеток до 4,58±1,62 пг/(клетка×сут). При культивировании лидерного клона-продуцента, коэкспрессирующего арилсульфатазу В и формилглицин-генерирующий фермент, в условиях периодического культивирования с подпиткой длительностью 12 сут достигнуто увеличение выхода активного лизосомального фермента арилсульфатазы В до 420 мг/л при добавлении в ростовую среду сульфата меди в концентрации 300 мкM. Выводы: культивирование клонов-продуцентов, коэкспрессирующих арилсульфатазу В и формилглицин-генерирующий фермент, в условиях периодического культивирования с подпиткой и с добавлением в среду сульфата меди приводит к значительному улучшению ростовых свойств клеточной линии и выхода целевого фермента. Данный подход в подборе условий культивирования продуцентов можно применять к другим ферментам подкласса сульфатаз

    Development and Validation of a Biolayer Interferometry Method for Determination of Human Anti-PD-1 Monoclonal Antibody Concentration in Cynomolgus Serum

    Get PDF
    Some types of immunotherapy of malignant tumours are aimed at restoration of T-cells’ ability to recognize and eliminate cancer. Programmed cell death ligand-1 (PD-L1) overexpression is characteristic of many human tumours and is associated with poor prognosis for patients. The development of monoclonal antibodies (mAbs) specific for PD-L1 or PD-1 is a promising area of immunotherapy of malignant tumours. However, before a therapeutic antibody-based product enters the market, it is necessary to ensure its safety and efficacy, i.e. perform a full scope of preclinical and clinical studies. The aim of the study was to develop and validate a bioanalytical method that does not require additional labeling and that could be used for determination of mAbs specific for human PD-L1 in the blood serum of a biological test system during preclinical studies. Materials and methods: an antigen in the form of a dimer of PD-1 extra-cellular domain covalently bonded to the Fc-fragment of human IgG (R&D Systems, USA) was used in the study. The antigen was immobilised on Dip and Read™ Protein A biosensors (Fortebio, USA). The therapeutic anti-PD-1 antibody GNR-051 was developed and produced by IBC “Generium” (Russia). The healthy cynomolgus monkey serum samples used as matrix were obtained from the Research Institute of Medical Primatology (Sochi, Russia). The assessment of binding was performed using Octet® QKe interferometer (Fortebio, USA) by real-time analysis of the dose-dependent rate of the antigen-antibody complex formation. Results: the paper presents experimental data on the development and validation of the test method for determination of the therapeutic PD-1-binding mAb concentration in cynomolgus monkey serum in the antibody concentration range from 2 to 2500 µg/mL. The authors assessed the calibration curve reliability, between-run and within-run precision and accuracy, dilution linearity, specificity and selectivity of the test method. Conclusions: the authors developed and validated the biolayer interferometry-based method for determination of therapeutic mAbs concentration. The method was shown to comply with the Eurasian Economic Union’s regulatory requirements in terms of the main validation parameters: analytical range, accuracy, precision, and selectivity

    The novel highly effective ir-photosensitizer for photodynamic therapy of cancer

    No full text
    The present article is devoted to the synthesis and investigation of biological and physico-chemical properties of the bacteriopurpurinimide derivative that was used as a photosensitizer. These substances have an ability to accumulate selectively in the malignant tumours. Also this class of photosensitizers is of particular interest because of their strong light absorbtion spectrum in red and NIR-region in the “window of tissue transparency”. This property affords to treat deep-seated tumours at the depth of 2 cm. In this study it was implemented the method of photodynamic therapy of cancer. The photosensitizer for this study was prepared as the nanostructured water-soluble micellar dispersion of O-propyloxime-N-propoxybacteriopurpurinimide methyl ester. The photodynamic therapy using this substance provided the highly effective photoinduced antitumour in vitro and in vivo activity, 100% regression of the tumour and 90% cure of animals

    Internalization of Recombinant Imiglucerase into Mouse Peritoneal Macrophages and L929 Mouse Fibroblasts

    Get PDF
    Enzyme replacement therapy (ERT) is one of the most efficient treatments for lysosomal storage diseases. Type 1 Gaucher disease is caused by β-glucocerebrosidase enzyme deficiency, which may be compensated for by intravenous infusions of imiglucerase—a recombinant enzyme. Imiglucerase targets macrophages and enters these cells via interaction with mannose receptors on the cell membrane. Characterisation of internalization of enzymes by target cells is important in the context of the development of new medicines and production of existing ERT medicines. The peritoneal and alveolar macrophages, as well as macrophages of the spleen of small laboratory animals (rats and mice) are widely used in such studies. However, isolation of cells from animal sources raises ethical issues, and therefore continuous mammalian cell lines may offer an attractive alternative. The aim of the study: to conduct comparative studies on the internalization of recombinant imiglucerase into mouse peritoneal macrophages and L929 mouse fibroblasts. Materials and methods: CerezymeR batches 7HV0913, C6214H05, 7HV0888 (Genzyme Ltd., UK); Glurazim batches 020416, 011117, 021117 (LLC “IBC “Generium”, Russia). We used peritoneal macrophages obtained from BALB/c mice and L929 mouse fibroblasts. The cells were cultured in DMEM/F12 complete growth medium with 10% fetal bovine serum. The activity of imiglucerase internalized into the cells was evaluated spectrophotometrically by hydrolysis of the artificial substrate—4-methylumbelliferyl-β-Dglucopyranoside. Results: the study compared internalization of recombinant imiglucerase (the active ingredient of CerezymeR and Glurazim) by mouse peritoneal macrophages and L929 mouse fibroblasts. It was demonstrated that the medicines activity in the lysates of peritoneal macrophages is comparable with that in the lysates of L929 mouse fibroblasts. Regardless of the model system, the activity of Glurazim stayed within the acceptable range (80–125%) established for biosimilar products. Conclusions: the experiments proved that L929 mouse fibroblasts could be recommended for assessment of internalization of recombinant imiglucerase

    Results of a phase I open randomized comparative crossover clinical trial to assess the safety and pharmacokinetics of Glurazyme® (imiglucerase) in comparison with the reference product in healthy volunteers

    Get PDF
    Background. Currently, the main treatment for Gaucher disease is enzyme replacement therapy. Recombinant glucocerebrosidase (imiglu-cerase) is the first biotechnological drug for enzyme replacement therapy with proven clinical efficacy and safety for the treatment of patients of different ages with Gaucher disease type 1 and type 3, used in clinical practice since 1994. In Russia, within the framework of the “Pharma 2020” pharmaceutical industry development strategy, the first biosimilar of imiglucerase, the drug Glurazyme®, was developed. The obtained results of preclinical studies became the basis for a phase I randomized comparative crossover clinical trial.The objective of the study was to assess the short-term safety and pharmacokinetic parameters of Glurazyme® in comparison with the Cerezyme® after a single intravenous administration to healthy volunteers.Materials and methods. 23 healthy volunteers aged 18—45 years were included in a 3-stage clinical trial. The study during the 1st and 2nd stages was open, randomized, comparative, crossover. At the 1st stage, volunteers from the 1st group received the Glurazyme®, from the 2nd group — the Cerezyme® once in doses of 30 U/kg. At the 2nd stage, Cerezyme® was administered to the 1st group, Glurazyme® - to the 2nd group once at doses of 30 U/kg. After the end of the 1st and 2nd stages, the 3rd stage was carried out for the 3rd group (n = 5) with the administration of the test drug once at a dose of 60 U/kg.Results. For all studied pharmacokinetic parameters, after administration of the test and reference drugs in doses of 30 U/kg, 90 % confidence interval was in the range from 80 to 125 %, which indicates the pharmacokinetic compared drugs equivalence. A total of 6 adverse events of mild and moderate severity were recorded. Of these, 4 adverse events were noted after administration of the study drug and were not associated with its administration. A comparative analysis of safety assessment parameters in this study (frequency and severity of adverse events, physical examination of healthy volunteers with an assessment of vital signs, laboratory tests, electrocardiography) did not reveal intergroup differences.Conclusion. The pharmacokinetic equivalence of the Glurazyme® and the reference drug in a dose of 30 U/kg has been established. A nonlinear dependence of the main pharmacokinetic parameters on studied drug administered dose was revealed. Safety and the absence of adverse reactions after a single injection of the study drug are shown

    Разработка и валидация методики определения концентрации антитела человека, блокирующего связывание PD-1 с лигандами, в сыворотке крови яванского макака методом биослойной интерферометрии

    Get PDF
    Some types of immunotherapy of malignant tumours are aimed at restoration of T-cells’ ability to recognize and eliminate cancer. Programmed cell death ligand-1 (PD-L1) overexpression is characteristic of many human tumours and is associated with poor prognosis for patients. The development of monoclonal antibodies (mAbs) specific for PD-L1 or PD-1 is a promising area of immunotherapy of malignant tumours. However, before a therapeutic antibody-based product enters the market, it is necessary to ensure its safety and efficacy, i.e. perform a full scope of preclinical and clinical studies. The aim of the study was to develop and validate a bioanalytical method that does not require additional labeling and that could be used for determination of mAbs specific for human PD-L1 in the blood serum of a biological test system during preclinical studies. Materials and methods: an antigen in the form of a dimer of PD-1 extra-cellular domain covalently bonded to the Fc-fragment of human IgG (R&D Systems, USA) was used in the study. The antigen was immobilised on Dip and Read™ Protein A biosensors (Fortebio, USA). The therapeutic anti-PD-1 antibody GNR-051 was developed and produced by IBC “Generium” (Russia). The healthy cynomolgus monkey serum samples used as matrix were obtained from the Research Institute of Medical Primatology (Sochi, Russia). The assessment of binding was performed using Octet® QKe interferometer (Fortebio, USA) by real-time analysis of the dose-dependent rate of the antigen-antibody complex formation. Results: the paper presents experimental data on the development and validation of the test method for determination of the therapeutic PD-1-binding mAb concentration in cynomolgus monkey serum in the antibody concentration range from 2 to 2500 µg/mL. The authors assessed the calibration curve reliability, between-run and within-run precision and accuracy, dilution linearity, specificity and selectivity of the test method. Conclusions: the authors developed and validated the biolayer interferometry-based method for determination of therapeutic mAbs concentration. The method was shown to comply with the Eurasian Economic Union’s regulatory requirements in terms of the main validation parameters: analytical range, accuracy, precision, and selectivity. Целью некоторых видов иммунотерапии злокачественных опухолей является восстановление способности Т-клеток распознавать и уничтожать опухолевые клетки. Сверхэкспрессия лиганда запрограммированной клеточной смерти (PD-L1) распространена во многих опухолях человека и связана с плохим прогнозом для пациента. Разработка моноклональных антител, специфичных к PD-L1 или PD-1, является перспективным направлением в иммунотерапии злокачественных новообразований. Однако прежде чем лекарственный препарат на основе терапевтического антитела появится на фармацевтическом рынке, необходимо убедиться в его безопасности и эффективности, то есть провести доклинические и клинические исследования в полном объеме. Цель работы: разработать и валидировать биоаналитическую методику, позволяющую без внесения дополнительных меток определить концентрацию моноклонального антитела, специфичного к PD-1 человека, в сыворотке крови биологической тест-системы в ходе проведения доклинических исследований. Материалы и методы: в работе использовали антиген в виде димера внеклеточного домена рецептора PD-1, ковалентно связанного с Fc-фрагментом иммуноглобулина человека (R&D Systems, США). Антиген иммобилизовали на биосенсоры Dip and Read™ Protein A (Fortebio, США). Терапевтическое моноклональное анти-PD-1 антитело GNR-051 произведено в ООО «МБЦ «Генериум» (Россия). Используемые в качестве матрицы образцы сыворотки крови здоровых яванских макак получены в ФГБНУ «НИИ МП» (г. Сочи, Россия). Оценку связывания проводили с помощью интерферометра Octet® QKe (Fortebio, США) в режиме наблюдения в реальном времени дозозависимой скорости формирования комплекса антиген–антитело. Результаты: представлены экспериментальные данные по разработке и валидации методики определения концентрации препарата моноклонального терапевтического антитела, связывающегося с клеточным рецептором PD-1, в сыворотке крови яванского макака в диапазоне концентраций антитела 2–2500 мкг/мл. Проведена оценка устойчивости параметров градуировочной кривой, оценка правильности и прецизионности между опытами и внутри опыта, оценка линейности разведения, исследована специфичность и селективность методики. Выводы: разработана и валидирована методика определения концентрации препарата моноклонального терапевтического антитела на основе биослойной интерферометрии. Показано соответствие методики требованиям нормативных документов ЕврАзЭС по основным валидационным характеристикам – аналитическому диапазону, правильности, прецизионности и селективности
    corecore