Risk factors and comorbidities associated with magnesium deficiency in pregnant women and women with hormone-related conditions: analysis of a large real-world dataset
|
01.12.2021 |
Orlova S.
Dikke G.
Pickering G.
Yaltseva N.
Konchits S.
Starostin K.
Bevz A.
|
BMC Pregnancy and Childbirth |
10.1186/s12884-021-03558-2 |
0 |
Ссылка
© 2021, The Author(s). Background: An accumulating body of literature indicates that magnesium deficiency is associated with a number of hormone-related conditions (HRC) in women, and epidemiological studies are needed to assess its prevalence and risk factors. Here, we present a secondary analysis of data pooled from four large observational studies that assessed magnesium deficiency among pregnant women and women with HRC across the Russian Federation. Methods: The main objective of this analysis was to estimate the prevalence of magnesium deficiency in this population and to describe risk factors and comorbidities associated with low serum magnesium. Univariate logistic regression analysis was performed to identify the risk factors and comorbid conditions associated with an increased risk of low serum magnesium level. Results: A total of 983 pregnant women and 9444 women with HRC were eligible for analysis. Prevalence of hypomagnesemia (magnesium serum level cut-off < 0.66 mmol/L/< 0.8 mmol/L) was 34.0%/78.9% in pregnant women and 21.4%/54.8% in women with HRC. The highest prevalence of magnesium deficiency was observed for osteoporosis and climacteric syndrome. Risk factors included diastolic blood pressure, previous pregnancy complications, infections and edema for pregnant women, and age, body mass index, and various comorbidities for women with HRC. Conclusions: These results confirm the high prevalence of hypomagnesemia in pregnant women and women with HRC and underline the importance of routine screening, since risk factors are mostly non-specific.
Читать
тезис
|
CAR T cells in solid tumors: challenges and opportunities
|
01.12.2021 |
Marofi F.
Motavalli R.
Safonov V.A.
Thangavelu L.
Yumashev A.V.
Alexander M.
Shomali N.
Chartrand M.S.
Pathak Y.
Jarahian M.
Izadi S.
Hassanzadeh A.
Shirafkan N.
Tahmasebi S.
Khiavi F.M.
|
Stem Cell Research and Therapy |
10.1186/s13287-020-02128-1 |
0 |
Ссылка
© 2021, The Author(s). Background: CARs are simulated receptors containing an extracellular single-chain variable fragment (scFv), a transmembrane domain, as well as an intracellular region of immunoreceptor tyrosine-based activation motifs (ITAMs) in association with a co-stimulatory signal. Main body: Chimeric antigen receptor (CAR) T cells are genetically engineered T cells to express a receptor for the recognition of the particular surface marker that has given rise to advances in the treatment of blood disorders. The CAR T cells obtain supra-physiological properties and conduct as “living drugs” presenting both immediate and steady effects after expression in T cells surface. But, their efficacy in solid tumor treatment has not yet been supported. The pivotal challenges in the field of solid tumor CAR T cell therapy can be summarized in three major parts: recognition, trafficking, and surviving in the tumor. On the other hand, the immunosuppressive tumor microenvironment (TME) interferes with T cell activity in terms of differentiation and exhaustion, and as a result of the combined use of CARs and checkpoint blockade, as well as the suppression of other inhibitor factors in the microenvironment, very promising results were obtained from the reduction of T cell exhaustion. Conclusion: Nowadays, identifying and defeating the mechanisms associated with CAR T cell dysfunction is crucial to establish CAR T cells that can proliferate and lyse tumor cells severely. In this review, we discuss the CAR signaling and efficacy T in solid tumors and evaluate the most significant barriers in this process and describe the most novel therapeutic methods aiming to the acquirement of the promising therapeutic outcome in non-hematologic malignancies.
Читать
тезис
|
Risk factors and comorbidities associated with magnesium deficiency in pregnant women and women with hormone-related conditions: analysis of a large real-world dataset
|
01.12.2021 |
Orlova S.
Dikke G.
Pickering G.
Yaltseva N.
Konchits S.
Starostin K.
Bevz A.
|
BMC Pregnancy and Childbirth |
10.1186/s12884-021-03558-2 |
0 |
Ссылка
© 2021, The Author(s). Background: An accumulating body of literature indicates that magnesium deficiency is associated with a number of hormone-related conditions (HRC) in women, and epidemiological studies are needed to assess its prevalence and risk factors. Here, we present a secondary analysis of data pooled from four large observational studies that assessed magnesium deficiency among pregnant women and women with HRC across the Russian Federation. Methods: The main objective of this analysis was to estimate the prevalence of magnesium deficiency in this population and to describe risk factors and comorbidities associated with low serum magnesium. Univariate logistic regression analysis was performed to identify the risk factors and comorbid conditions associated with an increased risk of low serum magnesium level. Results: A total of 983 pregnant women and 9444 women with HRC were eligible for analysis. Prevalence of hypomagnesemia (magnesium serum level cut-off < 0.66 mmol/L/< 0.8 mmol/L) was 34.0%/78.9% in pregnant women and 21.4%/54.8% in women with HRC. The highest prevalence of magnesium deficiency was observed for osteoporosis and climacteric syndrome. Risk factors included diastolic blood pressure, previous pregnancy complications, infections and edema for pregnant women, and age, body mass index, and various comorbidities for women with HRC. Conclusions: These results confirm the high prevalence of hypomagnesemia in pregnant women and women with HRC and underline the importance of routine screening, since risk factors are mostly non-specific.
Читать
тезис
|
CAR T cells in solid tumors: challenges and opportunities
|
01.12.2021 |
Marofi F.
Motavalli R.
Safonov V.A.
Thangavelu L.
Yumashev A.V.
Alexander M.
Shomali N.
Chartrand M.S.
Pathak Y.
Jarahian M.
Izadi S.
Hassanzadeh A.
Shirafkan N.
Tahmasebi S.
Khiavi F.M.
|
Stem Cell Research and Therapy |
10.1186/s13287-020-02128-1 |
0 |
Ссылка
© 2021, The Author(s). Background: CARs are simulated receptors containing an extracellular single-chain variable fragment (scFv), a transmembrane domain, as well as an intracellular region of immunoreceptor tyrosine-based activation motifs (ITAMs) in association with a co-stimulatory signal. Main body: Chimeric antigen receptor (CAR) T cells are genetically engineered T cells to express a receptor for the recognition of the particular surface marker that has given rise to advances in the treatment of blood disorders. The CAR T cells obtain supra-physiological properties and conduct as “living drugs” presenting both immediate and steady effects after expression in T cells surface. But, their efficacy in solid tumor treatment has not yet been supported. The pivotal challenges in the field of solid tumor CAR T cell therapy can be summarized in three major parts: recognition, trafficking, and surviving in the tumor. On the other hand, the immunosuppressive tumor microenvironment (TME) interferes with T cell activity in terms of differentiation and exhaustion, and as a result of the combined use of CARs and checkpoint blockade, as well as the suppression of other inhibitor factors in the microenvironment, very promising results were obtained from the reduction of T cell exhaustion. Conclusion: Nowadays, identifying and defeating the mechanisms associated with CAR T cell dysfunction is crucial to establish CAR T cells that can proliferate and lyse tumor cells severely. In this review, we discuss the CAR signaling and efficacy T in solid tumors and evaluate the most significant barriers in this process and describe the most novel therapeutic methods aiming to the acquirement of the promising therapeutic outcome in non-hematologic malignancies.
Читать
тезис
|
Mesenchymal stem cells and cancer therapy: insights into targeting the tumour vasculature
|
01.12.2021 |
Aravindhan S.
Ejam S.S.
Lafta M.H.
Markov A.
Yumashev A.V.
Ahmadi M.
|
Cancer Cell International |
10.1186/s12935-021-01836-9 |
0 |
Ссылка
© 2021, The Author(s). A crosstalk established between tumor microenvironment and tumor cells leads to contribution or inhibition of tumor progression. Mesenchymal stem cells (MSCs) are critical cells that fundamentally participate in modulation of the tumor microenvironment, and have been reported to be able to regulate and determine the final destination of tumor cell. Conflicting functions have been attributed to the activity of MSCs in the tumor microenvironment; they can confer a tumorigenic or anti-tumor potential to the tumor cells. Nonetheless, MSCs have been associated with a potential to modulate the tumor microenvironment in favouring the suppression of cancer cells, and promising results have been reported from the preclinical as well as clinical studies. Among the favourable behaviours of MSCs, are releasing mediators (like exosomes) and their natural migrative potential to tumor sites, allowing efficient drug delivering and, thereby, efficient targeting of migrating tumor cells. Additionally, angiogenesis of tumor tissue has been characterized as a key feature of tumors for growth and metastasis. Upon introduction of first anti-angiogenic therapy by a monoclonal antibody, attentions have been drawn toward manipulation of angiogenesis as an attractive strategy for cancer therapy. After that, a wide effort has been put on improving the approaches for cancer therapy through interfering with tumor angiogenesis. In this article, we attempted to have an overview on recent findings with respect to promising potential of MSCs in cancer therapy and had emphasis on the implementing MSCs to improve them against the suppression of angiogenesis in tumor tissue, hence, impeding the tumor progression.
Читать
тезис
|
Mesenchymal stem cells and cancer therapy: insights into targeting the tumour vasculature
|
01.12.2021 |
Aravindhan S.
Ejam S.S.
Lafta M.H.
Markov A.
Yumashev A.V.
Ahmadi M.
|
Cancer Cell International |
10.1186/s12935-021-01836-9 |
0 |
Ссылка
© 2021, The Author(s). A crosstalk established between tumor microenvironment and tumor cells leads to contribution or inhibition of tumor progression. Mesenchymal stem cells (MSCs) are critical cells that fundamentally participate in modulation of the tumor microenvironment, and have been reported to be able to regulate and determine the final destination of tumor cell. Conflicting functions have been attributed to the activity of MSCs in the tumor microenvironment; they can confer a tumorigenic or anti-tumor potential to the tumor cells. Nonetheless, MSCs have been associated with a potential to modulate the tumor microenvironment in favouring the suppression of cancer cells, and promising results have been reported from the preclinical as well as clinical studies. Among the favourable behaviours of MSCs, are releasing mediators (like exosomes) and their natural migrative potential to tumor sites, allowing efficient drug delivering and, thereby, efficient targeting of migrating tumor cells. Additionally, angiogenesis of tumor tissue has been characterized as a key feature of tumors for growth and metastasis. Upon introduction of first anti-angiogenic therapy by a monoclonal antibody, attentions have been drawn toward manipulation of angiogenesis as an attractive strategy for cancer therapy. After that, a wide effort has been put on improving the approaches for cancer therapy through interfering with tumor angiogenesis. In this article, we attempted to have an overview on recent findings with respect to promising potential of MSCs in cancer therapy and had emphasis on the implementing MSCs to improve them against the suppression of angiogenesis in tumor tissue, hence, impeding the tumor progression.
Читать
тезис
|
Any closer to successful therapy of multiple myeloma? CAR-T cell is a good reason for optimism
|
01.12.2021 |
Marofi F.
Tahmasebi S.
Rahman H.S.
Kaigorodov D.
Markov A.
Yumashev A.V.
Shomali N.
Chartrand M.S.
Pathak Y.
Mohammed R.N.
Jarahian M.
Motavalli R.
Motavalli Khiavi F.
|
Stem Cell Research and Therapy |
10.1186/s13287-021-02283-z |
0 |
Ссылка
Despite many recent advances on cancer novel therapies, researchers have yet a long way to cure cancer. They have to deal with tough challenges before they can reach success. Nonetheless, it seems that recently developed immunotherapy-based therapy approaches such as adoptive cell transfer (ACT) have emerged as a promising therapeutic strategy against various kinds of tumors even the cancers in the blood (liquid cancers). The hematological (liquid) cancers are hard to be targeted by usual cancer therapies, for they do not form localized solid tumors. Until recently, two types of ACTs have been developed and introduced; tumor-infiltrating lymphocytes (TILs) and chimeric antigen receptor (CAR)-T cells which the latter is the subject of our discussion. It is interesting about engineered CAR-T cells that they are genetically endowed with unique cancer-specific characteristics, so they can use the potency of the host immune system to fight against either solid or liquid cancers. Multiple myeloma (MM) or simply referred to as myeloma is a type of hematological malignancy that affects the plasma cells. The cancerous plasma cells produce immunoglobulins (antibodies) uncontrollably which consequently damage the tissues and organs and break the immune system function. Although the last few years have seen significant progressions in the treatment of MM, still a complete remission remains unconvincing. MM is a medically challenging and stubborn disease with a disappointingly low rate of survival rate. When comparing the three most occurring blood cancers (i.e., lymphoma, leukemia, and myeloma), myeloma has the lowest 5-year survival rate (around 40%). A low survival rate indicates a high mortality rate with difficulty in treatment. Therefore, novel CAR-T cell-based therapies or combination therapies along with CAT-T cells may bring new hope for multiple myeloma patients. CAR-T cell therapy has a high potential to improve the remission success rate in patients with MM. To date, many preclinical and clinical trial studies have been conducted to investigate the ability and capacity of CAR T cells in targeting the antigens on myeloma cells. Despite the problems and obstacles, CAR-T cell experiments in MM patients revealed a robust therapeutic potential. However, several factors might be considered during CAR-T cell therapy for better response and reduced side effects. Also, incorporating the CAT-T cell method into a combinational treatment schedule may be a promising approach. In this paper, with a greater emphasis on CAR-T cell application in the treatment of MM, we will discuss and introduce CAR-T cell’s history and functions, their limitations, and the solutions to defeat the limitations and different types of modifications on CAR-T cells.
Читать
тезис
|
Renaissance of armored immune effector cells, CAR-NK cells, brings the higher hope for successful cancer therapy
|
01.12.2021 |
Marofi F.
Rahman H.S.
Thangavelu L.
Dorofeev A.
Bayas-Morejón F.
Shirafkan N.
Shomali N.
Chartrand M.S.
Jarahian M.
Vahedi G.
Mohammed R.N.
Shahrokh S.
Akbari M.
Khiavi F.M.
|
Stem Cell Research and Therapy |
10.1186/s13287-021-02251-7 |
0 |
Ссылка
In recent decades, a new method of cellular immunotherapy was introduced based on engineering and empowering the immune effector cells. In this type of immunotherapy, the immune effector cells are equipped with chimeric antigen receptor (CAR) to specifically target cancer cells. In much of the trials and experiments, CAR-modified T cell immunotherapy has achieved very promising therapeutic results in the treatment of some types of cancers and infectious diseases. However, there are also some considerable drawbacks in the clinical application of CAR-T cells although much effort is in progress to rectify the issues. In some conditions, CAR-T cells initiate over-activated and strong immune responses, therefore, causing unexpected side-effects such as systemic cytokine toxicity (i.e., cytokine release syndrome), neurotoxicity, on-target, off-tumor toxicity, and graft-versus-host disease (GvHD). To overcome these limitations in CAR-T cell immunotherapy, NK cells as an alternative source of immune effector cells have been utilized for CAR-engineering. Natural killer cells are key players of the innate immune system that can destroy virus-infected cells, tumor cells, or other aberrant cells with their efficient recognizing capability. Compared to T cells, CAR-transduced NK cells (CAR-NK) have several advantages, such as safety in clinical use, non-MHC-restricted recognition of tumor cells, and renewable and easy cell sources for their preparation. In this review, we will discuss the recent preclinical and clinical studies, different sources of NK cells, transduction methods, possible limitations and challenges, and clinical considerations.
Читать
тезис
|
CAR-engineered NK cells; a promising therapeutic option for treatment of hematological malignancies
|
01.12.2021 |
Marofi F.
Saleh M.M.
Rahman H.S.
Suksatan W.
Al-Gazally M.E.
Abdelbasset W.K.
Thangavelu L.
Yumashev A.V.
Hassanzadeh A.
Yazdanifar M.
Motavalli R.
Pathak Y.
Naimi A.
Baradaran B.
Nikoo M.
Khiavi F.M.
|
Stem Cell Research and Therapy |
10.1186/s13287-021-02462-y |
0 |
Ссылка
Adoptive cell therapy has received a great deal of interest in the treatment of advanced cancers that are resistant to traditional therapy. The tremendous success of chimeric antigen receptor (CAR)-engineered T (CAR-T) cells in the treatment of cancer, especially hematological cancers, has exposed CAR’s potential. However, the toxicity and significant limitations of CAR-T cell immunotherapy prompted research into other immune cells as potential candidates for CAR engineering. NK cells are a major component of the innate immune system, especially for tumor immunosurveillance. They have a higher propensity for immunotherapy in hematologic malignancies because they can detect and eliminate cancerous cells more effectively. In comparison to CAR-T cells, CAR-NK cells can be prepared from allogeneic donors and are safer with a lower chance of cytokine release syndrome and graft-versus-host disease, as well as being a more efficient antitumor activity with high efficiency for off-the-shelf production. Moreover, CAR-NK cells may be modified to target various antigens while also increasing their expansion and survival in vivo. Extensive preclinical research has shown that NK cells can be effectively engineered to express CARs with substantial cytotoxic activity against both hematological and solid tumors, establishing evidence for potential clinical trials of CAR-NK cells. In this review, we discuss recent advances in CAR-NK cell engineering in a variety of hematological malignancies, as well as the main challenges that influence the outcomes of CAR-NK cell-based tumor immunotherapies.
Читать
тезис
|
Novel CAR T therapy is a ray of hope in the treatment of seriously ill AML patients
|
01.12.2021 |
Marofi F.
Rahman H.S.
Al-Obaidi Z.M.J.
Jalil A.T.
Abdelbasset W.K.
Suksatan W.
Dorofeev A.E.
Shomali N.
Chartrand M.S.
Pathak Y.
Hassanzadeh A.
Baradaran B.
Ahmadi M.
Saeedi H.
Tahmasebi S.
Jarahian M.
|
Stem Cell Research and Therapy |
10.1186/s13287-021-02420-8 |
0 |
Ссылка
Acute myeloid leukemia (AML) is a serious, life-threatening, and hardly curable hematological malignancy that affects the myeloid cell progenies and challenges patients of all ages but mostly occurs in adults. Although several therapies are available including chemotherapy, allogeneic hematopoietic stem cell transplantation (alloHSCT), and receptor-antagonist drugs, the 5-year survival of patients is quietly disappointing, less than 30%. alloHSCT is the major curative approach for AML with promising results but the treatment has severe adverse effects such as graft-versus-host disease (GVHD). Therefore, as an alternative, more efficient and less harmful immunotherapy-based approaches such as the adoptive transferring T cell therapy are in development for the treatment of AML. As such, chimeric antigen receptor (CAR) T cells are engineered T cells which have been developed in recent years as a breakthrough in cancer therapy. Interestingly, CAR T cells are effective against both solid tumors and hematological cancers such as AML. Gradually, CAR T cell therapy found its way into cancer therapy and was widely used for the treatment of hematologic malignancies with successful results particularly with somewhat better results in hematological cancer in comparison to solid tumors. The AML is generally fatal, therapy-resistant, and sometimes refractory disease with a disappointing low survival rate and weak prognosis. The 5-year survival rate for AML is only about 30%. However, the survival rate seems to be age-dependent. Novel CAR T cell therapy is a light at the end of the tunnel. The CD19 is an important target antigen in AML and lymphoma and the CAR T cells are engineered to target the CD19. In addition, a lot of research goes on the discovery of novel target antigens with therapeutic efficacy and utilizable for generating CAR T cells against various types of cancers. In recent years, many pieces of research on screening and identification of novel AML antigen targets with the goal of generation of effective anti-cancer CAR T cells have led to new therapies with strong cytotoxicity against cancerous cells and impressive clinical outcomes. Also, more recently, an improved version of CAR T cells which were called modified or smartly reprogrammed CAR T cells has been designed with less unwelcome effects, less toxicity against normal cells, more safety, more specificity, longer persistence, and proliferation capability. The purpose of this review is to discuss and explain the most recent advances in CAR T cell-based therapies targeting AML antigens and review the results of preclinical and clinical trials. Moreover, we will criticize the clinical challenges, side effects, and the different strategies for CAR T cell therapy.
Читать
тезис
|
Challenging anticoagulation cases: Cancer-associated venous thromboembolism and chemotherapy-induced thrombocytopenia – A case-based review of clinical management
|
01.03.2021 |
Moik F.
Makatsariya A.
Ay C.
|
Thrombosis Research |
10.1016/j.thromres.2020.12.016 |
0 |
Ссылка
© 2020 The Author(s) Patients with cancer undergoing chemotherapy are at risk of thrombocytopenia. The co-incidence of cancer-associated venous thromboembolism (VTE) and thrombocytopenia is a frequent complication in patients with cancer. Especially in certain tumour entities at high VTE risk, chemotherapeutic agents with myelosuppressive effects are part of the standard of care. The management of cancer-associated VTE in the setting of chemotherapy-induced thrombocytopenia is challenging, in the absence of evidence from high-quality studies. Thrombocytopenia is associated with both increased risk of recurrent VTE and risk of bleeding during anticoagulation. In this case-based concise review, we aimed at summarizing available literature and expert consensus guidance on the treatment of cancer-associated VTE in patients with chemotherapy-induced thrombocytopenia.
Читать
тезис
|
Codelivery of STAT3 and PD-L1 siRNA by hyaluronate-TAT trimethyl/thiolated chitosan nanoparticles suppresses cancer progression in tumor-bearing mice
|
01.02.2021 |
Bastaki S.
Aravindhan S.
Ahmadpour Saheb N.
Afsari Kashani M.
Dorofeev A.E.
Karoon Kiani F.
Jahandideh H.
Beigi Dargani F.
Aksoun M.
Nikkhoo A.
Masjedi A.
Mahmoodpoor A.
Ahmadi M.
Dolati S.
Namvar S.
Jadidi-Niaragh F.
|
Life Sciences |
10.1016/j.lfs.2020.118847 |
0 |
Ссылка
© 2020 Immunotherapy methods using potential tumor microenvironment modulators have elicited durable therapeutic responses in cancer treatment. Immune checkpoint molecule programmed cell death-ligand 1 (PD-L1) and oncogenic transcription factor STAT3 (signal transducer and activator of transcription-3) assigned as inhibitory targets of our study and particular delivery system designed to deliver small interfering RNAs (siRNAs) to silence the targeted genes. Generated trimethyl chitosan (TMC) and thiolated chitosan (TC) nanoparticles (NPs) conjugated with HIV-1-derived TAT peptide and HA (hyaluronic acid) exhibited eligible physicochemical characteristics, notable siRNA encapsulation, serum stability, non-toxicity, controlled siRNA release, and extensive cellular uptake by cancer cells. Dual inhibition with STAT3/PD-L1 siRNA-loaded HA-TAT-TMC-TC NPs led to promising results, including significant downregulation of PD-L1 and STAT3 genes, striking suppressive effects on proliferation, migration, and angiogenesis of breast and melanoma cancer cell lines, and restrained tumor growth in vivo. These findings infer the capability of HA-TAT-TMC-TC NPs containing STAT3/PD-L1 siRNAs as a novel tumor-suppressive candidate in cancer treatment.
Читать
тезис
|
The feasibility of Miltuximab®-IRDye700DX-mediated photoimmunotherapy of solid tumors
|
01.12.2020 |
Polikarpov D.M.
Campbell D.H.
Lund M.E.
Lu Y.
Lu Y.
Wu J.
Walsh B.J.
Zvyagin A.V.
Gillatt D.A.
|
Photodiagnosis and Photodynamic Therapy |
10.1016/j.pdpdt.2020.102064 |
0 |
Ссылка
© 2020 Elsevier B.V. Background: Photoimmunotherapy (PIT) is an emerging method of cancer treatment based on the use of a photosensitizer near-infrared dye IRDye700DX (IR700) conjugated to a monoclonal antibody. The antibody selectively delivers IR700 to cancer cells, which can then be killed after photoexcitation. Glypican-1 (GPC-1) is a novel target expressed specifically in malignant tumors. We aimed to investigate whether anti-GPC-1 antibody Miltuximab® (Glytherix Ltd., Sydney, Australia) can be conjugated with IR700 for PIT of solid tumors. Methods: The dye IR700 was conjugated with Miltuximab® and characterized by spectrophotometry and flow cytometry. Miltuximab®-IR700-mediated PIT was tested in prostate (DU-145), bladder (C3 and T-24), brain (U-87 and U-251) and ovarian (SKOV-3) cancer cell lines. After 1 h incubation with Miltuximab®-IR700, the cells were washed by PBS and illuminated using a 690-nm light-emitting diode. The viability of the cells was assessed by a CCK-8 viability kit 24 h later. Results: Miltuximab®-IR700-mediated PIT caused 67.3–92.3% reduction in viability of cells with medium-high GPC-1 expression and did not affect the viability of GPC-1-low cells. Cytotoxicity was attributed to the targeted binding of the conjugate with subsequent photoactivation, as the conjugate or light exposure alone had no effect on the cell viability. Miltuximab®-IR700 did not induce cytotoxicity in cells blocked by unconjugated Miltuximab®. Conclusions: PIT with Miltuximab®-IR700 appears to be highly specific and effective against GPC-1-expressing cancer cells, indicating that it holds promise for an effective and safe treatment of early stage solid tumors or as adjuvant therapy following surgical resection. These findings necessitate further investigation of PIT with Miltuximab®-IR700 in other GPC-1-expressing cancer cell lines in vitro and in vivo in xenograft tumor models.
Читать
тезис
|
The feasibility of Miltuximab®-IRDye700DX-mediated photoimmunotherapy of solid tumors
|
01.12.2020 |
Polikarpov D.M.
Campbell D.H.
Lund M.E.
Lu Y.
Lu Y.
Wu J.
Walsh B.J.
Zvyagin A.V.
Gillatt D.A.
|
Photodiagnosis and Photodynamic Therapy |
10.1016/j.pdpdt.2020.102064 |
0 |
Ссылка
© 2020 Elsevier B.V. Background: Photoimmunotherapy (PIT) is an emerging method of cancer treatment based on the use of a photosensitizer near-infrared dye IRDye700DX (IR700) conjugated to a monoclonal antibody. The antibody selectively delivers IR700 to cancer cells, which can then be killed after photoexcitation. Glypican-1 (GPC-1) is a novel target expressed specifically in malignant tumors. We aimed to investigate whether anti-GPC-1 antibody Miltuximab® (Glytherix Ltd., Sydney, Australia) can be conjugated with IR700 for PIT of solid tumors. Methods: The dye IR700 was conjugated with Miltuximab® and characterized by spectrophotometry and flow cytometry. Miltuximab®-IR700-mediated PIT was tested in prostate (DU-145), bladder (C3 and T-24), brain (U-87 and U-251) and ovarian (SKOV-3) cancer cell lines. After 1 h incubation with Miltuximab®-IR700, the cells were washed by PBS and illuminated using a 690-nm light-emitting diode. The viability of the cells was assessed by a CCK-8 viability kit 24 h later. Results: Miltuximab®-IR700-mediated PIT caused 67.3–92.3% reduction in viability of cells with medium-high GPC-1 expression and did not affect the viability of GPC-1-low cells. Cytotoxicity was attributed to the targeted binding of the conjugate with subsequent photoactivation, as the conjugate or light exposure alone had no effect on the cell viability. Miltuximab®-IR700 did not induce cytotoxicity in cells blocked by unconjugated Miltuximab®. Conclusions: PIT with Miltuximab®-IR700 appears to be highly specific and effective against GPC-1-expressing cancer cells, indicating that it holds promise for an effective and safe treatment of early stage solid tumors or as adjuvant therapy following surgical resection. These findings necessitate further investigation of PIT with Miltuximab®-IR700 in other GPC-1-expressing cancer cell lines in vitro and in vivo in xenograft tumor models.
Читать
тезис
|
The feasibility of Miltuximab®-IRDye700DX-mediated photoimmunotherapy of solid tumors
|
01.12.2020 |
Polikarpov D.M.
Campbell D.H.
Lund M.E.
Lu Y.
Lu Y.
Wu J.
Walsh B.J.
Zvyagin A.V.
Gillatt D.A.
|
Photodiagnosis and Photodynamic Therapy |
10.1016/j.pdpdt.2020.102064 |
0 |
Ссылка
© 2020 Elsevier B.V. Background: Photoimmunotherapy (PIT) is an emerging method of cancer treatment based on the use of a photosensitizer near-infrared dye IRDye700DX (IR700) conjugated to a monoclonal antibody. The antibody selectively delivers IR700 to cancer cells, which can then be killed after photoexcitation. Glypican-1 (GPC-1) is a novel target expressed specifically in malignant tumors. We aimed to investigate whether anti-GPC-1 antibody Miltuximab® (Glytherix Ltd., Sydney, Australia) can be conjugated with IR700 for PIT of solid tumors. Methods: The dye IR700 was conjugated with Miltuximab® and characterized by spectrophotometry and flow cytometry. Miltuximab®-IR700-mediated PIT was tested in prostate (DU-145), bladder (C3 and T-24), brain (U-87 and U-251) and ovarian (SKOV-3) cancer cell lines. After 1 h incubation with Miltuximab®-IR700, the cells were washed by PBS and illuminated using a 690-nm light-emitting diode. The viability of the cells was assessed by a CCK-8 viability kit 24 h later. Results: Miltuximab®-IR700-mediated PIT caused 67.3–92.3% reduction in viability of cells with medium-high GPC-1 expression and did not affect the viability of GPC-1-low cells. Cytotoxicity was attributed to the targeted binding of the conjugate with subsequent photoactivation, as the conjugate or light exposure alone had no effect on the cell viability. Miltuximab®-IR700 did not induce cytotoxicity in cells blocked by unconjugated Miltuximab®. Conclusions: PIT with Miltuximab®-IR700 appears to be highly specific and effective against GPC-1-expressing cancer cells, indicating that it holds promise for an effective and safe treatment of early stage solid tumors or as adjuvant therapy following surgical resection. These findings necessitate further investigation of PIT with Miltuximab®-IR700 in other GPC-1-expressing cancer cell lines in vitro and in vivo in xenograft tumor models.
Читать
тезис
|
Immunosuppressive therapy of biopsy proved immune-mediated lymphocytic myocarditis in the virus-negative and virus-positive patients.
|
01.11.2020 |
Blagova O.
Nedostup A.
Kogan E.
Zaitsev A.
Fomin V.
|
Cardiovascular Pathology |
10.1016/j.carpath.2020.107260 |
0 |
Ссылка
© 2020 Purpose: to study the effect of immunosupressive therapy (IST) in the virus-negative and virus-positive patients with immune-mediated myocarditis. Methods: in 60 patients (45 male, 46.7 ± 11.8 years, mean LV EDD, 6.7 ± 0.7 cm, EF 26.2 ± 9.1%) active/borderline myocarditis was verified by endomyocardial biopsy (n = 38), intraoperative biopsy (n = 10), examination of explanted heart (n = 3) and autopsy (n = 9). Indications for IST determined based on histological, immune activity. The follow-up was 19.0 [7.25; 40.25] months. Results: The viral genome in the myocardium was detected in 32 patients (V+ group), incl. parvovirus B19 in 23. The anti-heart antibody level was equally high in the V+ and V- patients. Antiviral therapy was administered in 24 patients. IST (in 22 V+ and 24 V- patients) include steroids (n = 40), hydroxychloroquine (n = 20), azathioprine (n = 21). The significant decrease of LV EDD (6.7 ± 0.7 to 6.4 ± 0.8), PAP (48.9 ± 15.5 to 39.4 ± 11.5 mm Hg, р<0,01), increase of EF (26.5 ± 0.9 to 36.0 ± 10.8), and lower lethality (23.9% and 64.3%; RR 0.37, 95% CI 0.19–0.71), p<0.01, were found only in IST group. Significant improvement due to IST were achieved not only in V-, but also in V+ patients. Conclusions: IST in patients with immune-mediated lymphocytic myocarditis is effective and is associated with lower lethality both in virus-negative and virus-positive patients.
Читать
тезис
|
The exudative-constrictive tuberculosis pericarditis diagnosed by toracoscopic biopsy
|
01.08.2020 |
Blagova O.V.
Alijeva I.N.
Nedostup A.V.
Senchihin P.V.
Parshin V.D.
Kogan E.A.
|
Journal of Clinical Tuberculosis and Other Mycobacterial Diseases |
10.1016/j.jctube.2020.100165 |
0 |
Ссылка
© 2020 The Authors Purpose: To present the problems and possibilities of diagnostic and treatment in a patient with resistant exudative-constrictive pericarditis. Methods: The male patient 31 y. was admitted to the clinic due to exudative pericarditis and arthritis of the left knee joint. His medical history periodic febrile fever with a cough, episodes of syncope and atrial fibrillation, treatment with antibiotics and corticosteroids with a temporary effect. Results: No data were received for systemic disease, hypothyroidism, tumors. With CT in both lungs, small areas of fibrosis and lymphadenopathy were identified. Pericardial sheets diffusely thickened. EchoCG shows one liter of pericardial fluid with fibrin. All tests for viruses and tuberculosis are negative. Thoracoscopy was performed. Morphological examination showed tuberculosis granulomas with caseous necrosis. The growth of mycobacteria of tuberculosis from sputum was obtained. Therapy included pyrazinamide, ethambutol, levofloxacin, prednisolone 20 mg / day. Ponce's disease regressed. Due to the increase of constriction, subtotal pericardectomy was performed. Conclusion: Tuberculosis is one of the real causes of pericarditis with massive effusion and an outcome in constriction. The negative results of all laboratory tests for tuberculosis do not exclude the diagnosis. It is necessary to use invasive morphological diagnosis, including thoracoscopic biopsy.
Читать
тезис
|
The exudative-constrictive tuberculosis pericarditis diagnosed by toracoscopic biopsy
|
01.08.2020 |
Blagova O.V.
Alijeva I.N.
Nedostup A.V.
Senchihin P.V.
Parshin V.D.
Kogan E.A.
|
Journal of Clinical Tuberculosis and Other Mycobacterial Diseases |
10.1016/j.jctube.2020.100165 |
0 |
Ссылка
© 2020 The Authors Purpose: To present the problems and possibilities of diagnostic and treatment in a patient with resistant exudative-constrictive pericarditis. Methods: The male patient 31 y. was admitted to the clinic due to exudative pericarditis and arthritis of the left knee joint. His medical history periodic febrile fever with a cough, episodes of syncope and atrial fibrillation, treatment with antibiotics and corticosteroids with a temporary effect. Results: No data were received for systemic disease, hypothyroidism, tumors. With CT in both lungs, small areas of fibrosis and lymphadenopathy were identified. Pericardial sheets diffusely thickened. EchoCG shows one liter of pericardial fluid with fibrin. All tests for viruses and tuberculosis are negative. Thoracoscopy was performed. Morphological examination showed tuberculosis granulomas with caseous necrosis. The growth of mycobacteria of tuberculosis from sputum was obtained. Therapy included pyrazinamide, ethambutol, levofloxacin, prednisolone 20 mg / day. Ponce's disease regressed. Due to the increase of constriction, subtotal pericardectomy was performed. Conclusion: Tuberculosis is one of the real causes of pericarditis with massive effusion and an outcome in constriction. The negative results of all laboratory tests for tuberculosis do not exclude the diagnosis. It is necessary to use invasive morphological diagnosis, including thoracoscopic biopsy.
Читать
тезис
|
Newly formulated 5% 5-aminolevulinic acid photodynamic therapy on Candida albicans
|
01.03.2020 |
Greco G.
Di Piazza S.
Chan J.
Zotti M.
Hanna R.
Gheno E.
Zekiy A.
Pasquale C.
De Angelis N.
Amaroli A.
|
Photodiagnosis and Photodynamic Therapy |
10.1016/j.pdpdt.2019.10.010 |
0 |
Ссылка
© 2019 Elsevier B.V. Background: A large number of systemic diseases can be linked to oral candida pathogenicity. The global trend of invasive candidiasis has increased progressively and is often accentuated by increasing Candida albicans resistance to the most common antifungal medications. Photodynamic therapy (PDT) is a promising therapeutic approach for oral microbial infections. A new formulation of 5-aminolevulinic acid (5%ALA) in a thermosetting gel (t) (5%ALA-PTt) was patented and recently has become available on the market. However, its antimicrobial properties, whether mediated or not by PDT, are not yet known. In this work we characterised them. Methods: We isolated a strain of C. albicans from plaques on the oral mucus membrane of an infected patient. Colonies of this strain were exposed for 1 24 h, to 5%ALA-PTt, 5%ALA-PTt buffered to pH 6.5 (the pH of the oral mucosa) (5%ALA-PTtb) or not exposed (control). The 1 h-exposed samples were also irradiated at a wavelength of 630 nm with 0.14 watts (W) and 0.37 W/cm2 for 7 min at a distance of <1 mm. Results and conclusion: The 5% ALA-PTt preparation was shown to be effective in reducing the growth of biofilm and inoculum of C. albicans. This effect seems to be linked to the intrinsic characteristics of 5%ALA-TPt, such acidic pH and the induction of free radical production. This outcome was significantly enhanced by the effect of PDT at relatively short incubation and irradiation times, which resulted in growth inhibition of both treated biofilm and inoculum by ∼80% and ∼95%, respectively.
Читать
тезис
|
Newly formulated 5% 5-aminolevulinic acid photodynamic therapy on Candida albicans
|
01.03.2020 |
Greco G.
Di Piazza S.
Chan J.
Zotti M.
Hanna R.
Gheno E.
Zekiy A.
Pasquale C.
De Angelis N.
Amaroli A.
|
Photodiagnosis and Photodynamic Therapy |
10.1016/j.pdpdt.2019.10.010 |
0 |
Ссылка
© 2019 Elsevier B.V. Background: A large number of systemic diseases can be linked to oral candida pathogenicity. The global trend of invasive candidiasis has increased progressively and is often accentuated by increasing Candida albicans resistance to the most common antifungal medications. Photodynamic therapy (PDT) is a promising therapeutic approach for oral microbial infections. A new formulation of 5-aminolevulinic acid (5%ALA) in a thermosetting gel (t) (5%ALA-PTt) was patented and recently has become available on the market. However, its antimicrobial properties, whether mediated or not by PDT, are not yet known. In this work we characterised them. Methods: We isolated a strain of C. albicans from plaques on the oral mucus membrane of an infected patient. Colonies of this strain were exposed for 1 24 h, to 5%ALA-PTt, 5%ALA-PTt buffered to pH 6.5 (the pH of the oral mucosa) (5%ALA-PTtb) or not exposed (control). The 1 h-exposed samples were also irradiated at a wavelength of 630 nm with 0.14 watts (W) and 0.37 W/cm2 for 7 min at a distance of <1 mm. Results and conclusion: The 5% ALA-PTt preparation was shown to be effective in reducing the growth of biofilm and inoculum of C. albicans. This effect seems to be linked to the intrinsic characteristics of 5%ALA-TPt, such acidic pH and the induction of free radical production. This outcome was significantly enhanced by the effect of PDT at relatively short incubation and irradiation times, which resulted in growth inhibition of both treated biofilm and inoculum by ∼80% and ∼95%, respectively.
Читать
тезис
|