Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Stem Cell Res Ther ; 15(1): 111, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38644499

ABSTRACT

BACKGROUND: Graft failure (GF) is a rare but serious complication after allogeneic hematopoietic stem cell transplantation (HSCT). Prevention of graft failure remains the most advisable approach as there is no clear recommendation for the best strategies for reversing this complication. Administration of growth factor, additional hematopoietic progenitor boost, or a salvage HSCT are current modalities recommended for the treatment of GF. Autologous recovery without evidence of disease relapse occurs rarely in patients with GF, and in the absence of autologous recovery, further salvage transplantation following a second conditioning regimen is a potential treatment option that offers the best chances of long-term disease-free survival. The preconditioning regimens of second HSCT have a significant impact on engraftment and outcome, however, currently there is no consensus on optimal conditioning regimen for second HSCT in patients who have developed GF. Furthermore, a second transplant from a different donor or the same donor is still a matter of debate. OBSERVATIONS: We present our experience in managing pediatric patients with acute leukemia who encountered graft failure following stem cell transplantation. CONCLUSIONS AND RELEVANCE: Although a second transplantation is almost the only salvage method, we illustrate that some pediatric patients with acute leukemia who experience graft failure after an allogeneic stem cell transplant using Myeloablative conditioning (MAC) regimen may achieve long-term disease-free survival through autologous hematopoiesis recovery.


Subject(s)
Hematopoietic Stem Cell Transplantation , Transplantation Conditioning , Humans , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cell Transplantation/adverse effects , Child , Female , Male , Transplantation Conditioning/methods , Child, Preschool , Transplantation, Homologous/methods , Adolescent , Graft Rejection , Acute Disease , Transplantation, Autologous , Infant , Leukemia, Myeloid, Acute/therapy
2.
Mol Biol Rep ; 51(1): 460, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38551770

ABSTRACT

BACKGROUND: Among neurological diseases, multiple sclerosis (MS) affects mostly young adults and can cause long-term disability. While most medications with approval from regulatory agencies are very effective in treating MS disease, they are unable to repair the tissue damage found in the central nervous system (CNS). Consequently, Cell-based therapy particularly using mesenchymal stem/stromal cells (MSCs), holds promise for neuroprotection and tissue repair in MS treatment. Furthermore, placenta-derived MSCs (PLMSCs) have shown the potential to treat MS due to their abundance, noninvasive isolation from discarded tissues, no ethical problems, anti-inflammatory, and reparative properties. Accordingly, good manufacturing practices (GMPs) plays a crucial part in clinical SCs manufacturing. The purpose of our article is to discuss GMP-grade PLMSC protocols for treating MS as well as other clinical applications. METHODS AND RESULTS: Placental tissue obtained of a healthy donor during the caesarean delivery and PLMSCs isolated by GMP standards. Flow cytometry was used to assess the expression of the CD markers CD34, CD105, CD90, and CD73 in the MSCs and the mesodermal differentiation ability was evaluated. Furthermore, Genetic evaluation of PLMSCs was done by G-banded karyotyping and revealed no chromosomal instability. In spite of the anatomical origin of the starting material, PLMSCs using this method of culture were maternal in origin. CONCLUSIONS: We hope that our protocol for clinical manufacturing of PLMSCs according to GMP standards will assist researchers in isolating MSCs from placental tissue for clinical and pre-clinical applications.


Subject(s)
Mesenchymal Stem Cells , Multiple Sclerosis , Young Adult , Humans , Female , Pregnancy , Multiple Sclerosis/therapy , Multiple Sclerosis/metabolism , Placenta , Mesenchymal Stem Cells/metabolism , Flow Cytometry , Cell- and Tissue-Based Therapy , Cells, Cultured , Cell Differentiation , Cell Proliferation
3.
Cells ; 13(2)2024 01 22.
Article in English | MEDLINE | ID: mdl-38275825

ABSTRACT

Unlike MCF-7 cells, MDA-MB-231 cells are unresponsive to hormone therapy and often show resistance to chemotherapy and radiotherapy. Here, the antiproliferative effect of biocompatible montmorillonite (Mt) nanosheets on MDA-MB-231 and MCF-7 human breast cancer cells was evaluated by MTT assay, flow cytometry, and qRT-PCR. The results showed that the Mt IC50 for MDA-MB-231 and MCF-7 cells in a fetal bovine serum (FBS)-free medium was ~50 and ~200 µg/mL, and in 10% FBS medium ~400 and ~2000 µg/mL, respectively. Mt caused apoptosis in both cells by regulating related genes including Cas-3, P53, and P62 in MDA-MB-231 cells and Bcl-2, Cas-8, Cas-9, P53, and P62 in MCF-7 cells. Also, Mt arrested MCF-7 cells in the G0/G1 phase by altering Cyclin-D1 and P21 expression, and caused sub-G1 arrest and necrosis in both cells, possibly through damaging the mitochondria. However, fewer gene expression changes and more sub-G1 arrest and necrosis were observed in MDA-MB-231 cells, confirming the higher vulnerability of MDA-MB-231 cells to Mt. Furthermore, MDA-MB-231 cells appeared to be much more vulnerable to Mt compared to other cell types, including normal lung fibroblast (MRC-5), colon cancer (HT-29), and liver cancer (HepG2) cells. The higher vulnerability of MDA-MB-231 cells to Mt was inferred to be due to their higher proliferation rate. Notably, Mt cytotoxicity was highly dependent on both the Mt concentration and serum level, which favors Mt for the local treatment of MDA-MB-231 cells. Based on these results, Mt can be considered as an antiproliferative nanoagent against MDA-MB-231 cells and may be useful in the development of local nanoparticle-based therapies.


Subject(s)
Bentonite , Breast Neoplasms , Humans , Female , MCF-7 Cells , Bentonite/pharmacology , Bentonite/metabolism , Cell Proliferation , Tumor Suppressor Protein p53/metabolism , Breast Neoplasms/drug therapy , Necrosis
4.
Immunotherapy ; 15(12): 963-973, 2023 08.
Article in English | MEDLINE | ID: mdl-37337708

ABSTRACT

NK cells are the first sentinels of the immune system that can recognize and eradicate transformed cells. Their activation without a need for additional signaling have attracted great attention on the use of NK cells as a promising option in cancer immunotherapy. However, the large-scale production of NK cells for successful NK cells therapy is a challenge that needs to be tackled. Engineering NK cells to avoid tumor escape and improve their antitumor potency are the other matters of focus that have widely been studied in the recent years. This paper reviews the most recent advances in the stem cell-derived NK cell technology and discusses the potential of the engineered NK cells for clinical applications in cancer immunotherapy.


NK cells are important cells in our body's defense system that can find and destroy tumor cells. These cells are made in bone marrow (in adults) or umbilical cord (in the embryonic period) from a population of cells called stem cells, and then released into the blood and lymph. Stem cells are the early ancestral cells that can differentiate into multiple cell types. Because the number and function of NK cells in a tumor context are reduced, thus we can use these stem cells to make lots of NK cells for treatment purposes. Scientists can also make these cells even better at killing tumors by changing them to have special sensors. In the end, NK cells are like superheroes that fight and kill tumor cells, and using stem cells to make them is a really promising way to help treat malignant diseases.


Subject(s)
Killer Cells, Natural , Neoplasms , Humans , Immunotherapy , Immunotherapy, Adoptive , Stem Cells , Neoplasms/therapy
5.
Int J MCH AIDS ; 11(2): e571, 2022.
Article in English | MEDLINE | ID: mdl-36466768

ABSTRACT

Polymerase chain reaction (PCR) for the detection of nucleic acids is the gold standard test for the diagnosis of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, there is the probability of false-negative results with this test, which poses a threat to public health. Here, we highlight some important factors that should be considered for reducing the false-negative results of the SARS-CoV-2 PCR test.

6.
Vet Med Sci ; 8(5): 2086-2091, 2022 09.
Article in English | MEDLINE | ID: mdl-35838746

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is the fifth most diagnosed cancer and the second leading cause of cancer-related deaths worldwide. Sorafenib is the standard treatment used in the advanced stages of HCC. Cell therapy with mesenchymal stem cells (MSCs)-based cell therapy has proven effective in immune regulation and tumour growth inhibition. OBJECTIVES: In this study, we investigated the anti-inflammatory effect of MSCs on HCC xenografts. METHODS: Human HepG2 cell lines were subcutaneously implanted into the flank of 12 nude mice, divided into three groups: the control group, the IV group (intravenous MSCs injection) and the local group (local MSCs injection). Mice were sacrificed 6 weeks after tumour implantation, and tumours were resected entirety. Quantitative real-time polymerase chain reaction (qRT-PCR) measured the gene expression of inflammatory markers, including tumour necrosis factor-α (TNF-α), interleukin (IL)-1α and IL-10. Aspartate transaminase (AST), alanine transaminase (ALT) and urea levels were measured using spectrophotometry to ensure the safety of MSC therapy. RESULTS: Gene expressions for all three inflammatory markers were reduced in both MSCs groups compared to the control group. AST, ALT and urea levels remained in normal ranges. CONCLUSIONS: MSC therapy can reduce inflammation in HCC xenograft mouse models.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Rodent Diseases , Alanine Transaminase/metabolism , Alanine Transaminase/pharmacology , Alanine Transaminase/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Aspartate Aminotransferases/metabolism , Aspartate Aminotransferases/pharmacology , Aspartate Aminotransferases/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/veterinary , Disease Models, Animal , Heterografts , Humans , Interleukin-10/metabolism , Interleukin-10/pharmacology , Interleukin-10/therapeutic use , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/veterinary , Mesenchymal Stem Cell Transplantation/veterinary , Mesenchymal Stem Cells/metabolism , Mice , Mice, Nude , Sorafenib/metabolism , Sorafenib/pharmacology , Sorafenib/therapeutic use , Tumor Necrosis Factor-alpha/metabolism
7.
Hum Cell ; 35(5): 1355-1363, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35796938

ABSTRACT

Long non-coding RNA (LncRNA) is a new type of non-coding RNA whose transcription is more than 200 nucleotides in length and can be up to 100 kb. The crucial regulatory function of lncRNAs in different cellular processes is now notable in many human diseases, especially in different steps of tumorigenesis, making them clinically significant. This research tried to collect all evidence obtained so far regarding Nuclear Receptor subfamily 2 group F member 2 Antisense RNA 1 (NR2F2-AS1) to explore its role in carcinogenesis and molecular mechanism in several cancers. Collecting evidence value an oncogenic role for NR2F2-AS1, whose dysregulation changes the status for cancerous cells to gain the supremacy toward cellular proliferation, dissemination, and ultimately migration. The NR2F2-AS1 acts as competitive endogenous RNA (ceRNA) and contains several microRNA response elements (MREs) for different microRNAs involved in various pathways such as PI3K/AKT, Wnt/ß-catenin, and TGF-ß. This clinically makes NR2F2-AS1 a remarkable lncRNA which contributes to cancer progression and invasion and perhaps could be a candidate as a prognostic marker or even a therapeutic target.


Subject(s)
MicroRNAs , RNA, Long Noncoding , COUP Transcription Factor II/genetics , COUP Transcription Factor II/metabolism , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Phosphatidylinositol 3-Kinases/metabolism , RNA, Antisense , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
8.
Int J Hematol Oncol Stem Cell Res ; 16(4): 250-263, 2022 Oct 01.
Article in English | MEDLINE | ID: mdl-36883106

ABSTRACT

Autophagy plays a critical role in balancing sources of energy in response to harsh conditions and nutrient deprivation. Autophagy allows cells to survive in harsh condition and also serve as a death mechanism. Any dysregulation in autophagy signaling may lead to several disorders. Autophagy has been proposed to explain chemotherapy resistance in acute myeloid leukemia (AML). This signaling pathway can either act as a tumor suppressive function or chemo-resistance mechanism. Conventional chemotherapy drugs enhance apoptosis and indicate clinical benefit, but in some cases, relapse and chemotherapy resistance are observed. In leukemia, autophagy may promote cell survival in response to chemotherapy drugs. Therefore, new strategies by inhibiting or activating autophagy may find a broad application for treating leukemia and may significantly enhance clinical outcomes. In this review, we discussed the dimensional role of autophagy in leukemia.

9.
Stem Cell Res Ther ; 12(1): 400, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34256857

ABSTRACT

Multiple sclerosis (MS) is a central nervous system (CNS) chronic illness with autoimmune, inflammatory, and neurodegenerative effects characterized by neurological disorder and axonal loss signs due to myelin sheath autoimmune T cell attacks. Existing drugs, including disease-modifying drugs (DMD), help decrease the intensity and frequency of MS attacks, inflammatory conditions, and CNS protection from axonal damage. As they cannot improve axonal repair and show side effects, new therapeutic options are required. In this regard, due to their neuroprotection properties, immunomodulatory effects, and the ability to differentiate into neurons, the transplantation of mesenchymal stromal cells (MSCs) can be used for MS therapy. The use of adipose-derived MSCs (AdMSCs) or autologous bone marrow MSCs (BMSCs) has demonstrated unexpected effects including the invasive and painful isolation method, inadequate amounts of bone marrow (BM) stem cells, the anti-inflammatory impact reduction of AdMSCs that are isolated from fat patients, and the cell number and differentiation potential decrease with an increase in the age of BMSCs donor. Researchers have been trying to search for alternate tissue sources for MSCs, especially fetal annexes, which could offer a novel therapeutic choice for MS therapy due to the limitation of low cell yield and invasive collection methods of autologous MSCs. The transplantation of MSCs for MS treatment is discussed in this review. Finally, it is suggested that allogeneic sources of MSCs are an appealing alternative to autologous MSCs and could hence be a potential novel solution to MS therapy.


Subject(s)
Hematopoietic Stem Cell Transplantation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Multiple Sclerosis , Pharmaceutical Preparations , Bone Marrow Cells , Humans , Multiple Sclerosis/therapy
10.
Stem Cell Res Ther ; 12(1): 213, 2021 03 29.
Article in English | MEDLINE | ID: mdl-33781314

ABSTRACT

BACKGROUND: Hepatic fibrosis is a common complication in transfusion-dependent thalassemia patients. Data on the co-transplantation of mesenchymal stem cells (MSCs) with hematopoietic stem cells (HSCs) in beta-thalassemia major patients are scarce. Therefore, we aimed to evaluate the effect of co-transplantation of bone marrow-derived MSC with HSCs on the liver fibrosis alleviation and transplant outcomes in class III beta-thalassemia major. METHODS: Between April 1998 and January 2017, a total of 224 consecutive patients with class III beta-thalassemia major underwent allogeneic HSCT in the Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran. To assess liver fibrotic changes after transplantation, 47 patients participated in the MSC plus HSC group and 30 patients in the HSC only group at the end of the follow-up period. All patients underwent laboratory tests, especially serum ferritin and liver function testing, hepatic T2* MRI, liver biopsy, and FibroScan before and 2 years after transplantation. Kaplan-Meier curves were derived to determine survival and were compared using the log-rank test. Repeated-measure, mixed-effect linear regression models were used to examine the changes in liver fibrosis over time. RESULTS: The 10-year OS rate was 71.84% in the mesenchymal group and 61.89% in the non-mesenchymal group (P value = 0.294), while the 10-year TFS rate was 63.64% in the mesenchymal group and 52.78% in the non-mesenchymal group (P value = 0.285). No significant difference was observed in the 10-year NRM, rejection rate, ANC engraftment, platelet engraftment, acute GvHD, and chronic GvHD between the two groups. In addition, the results of repeated-measure, mixed-effect linear regression models showed that none of the variables determining hepatic fibrosis had a significant difference between patients receiving MSCs and patients who did not receive MSCs. CONCLUSIONS: Based on the results of this study, a single infusion of MSCs at the time of HSCT to patients with class III beta-thalassemia major could not significantly improve the liver fibrosis alleviation and transplantation outcomes, including OS, TFS, TRM, rejection rate, ANC engraftment, platelet engraftment, acute GvHD, and chronic GvHD.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , beta-Thalassemia , Bone Marrow , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cells , Humans , Iran , Liver Cirrhosis/diagnostic imaging , Liver Cirrhosis/therapy , beta-Thalassemia/therapy
11.
IUBMB Life ; 73(1): 130-145, 2021 01.
Article in English | MEDLINE | ID: mdl-33205598

ABSTRACT

A little number of current autophagy inhibitors may have beneficial effects on the acute myeloid leukemia (AML) patients. However, there is a strong need to figure out which settings should be activated or inhibited in autophagy pathway to prevail drug resistance and also to improve current treatment options in leukemia. Therefore, this study aimed to compare the effects of well-known inhibitors of autophagy (as 3-MA, BafA1, and HCQ) in leukemia KG-1 and HL-60 cells exposed to arsenic trioxide (ATO) and/or all-trans retinoic acid (ATRA). Cell proliferation and cytotoxicity of cells were examined by MTT assay. Autophagy was studied by evaluating the development of acidic vesicular organelles, and the autophagosomes formation was investigated by acridine orange staining and transmission electron microscopy. Moreover, the gene and protein expressions levels of autophagy markers (ATGs, p62/SQSTM1, and LC-3B) were also performed by qPCR and western blotting, respectively. The rate of apoptosis and cell cycle were evaluated using flow cytometry. We compared the cytotoxic and apoptotic effects of ATO and/or ATRA in both cell lines and demonstrated that some autophagy markers upregulated in this context. Also, it was shown that autophagy blockers HCQ and/or BafA1 could potentiate the cytotoxic effects of ATO/ATRA, which were more pronounced in KG-1 cells compared to HL-60 cell line. This study showed the involvement of autophagy during the treatment of KG-1 and HL-60 cells by ATO/ATRA. This study proposed that therapy of ATO/ATRA in combination with HCQ can be considered as a more effective strategy for targeting leukemic KG-1 cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Autophagy , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Promyelocytic, Acute/drug therapy , Apoptosis , Arsenic Trioxide/administration & dosage , Cell Proliferation , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Leukemia, Promyelocytic, Acute/metabolism , Leukemia, Promyelocytic, Acute/pathology , Tretinoin/administration & dosage , Tumor Cells, Cultured
12.
J Cell Physiol ; 236(5): 4066-4075, 2021 05.
Article in English | MEDLINE | ID: mdl-33151570

ABSTRACT

Controlled-release drug delivery systems are promising platforms in medicine. Among various types of material in drug delivery, hydrogels are interesting ones. They are water-soluble and tissue compatible polymers with a high capacity to carry and release drugs in a controllable manner. In this study, we introduce the synthesis, characterization, and application of an α-amylase responsive hydrogel in controlled drug delivery. The newly synthesized starch-based hydrogels structurally characterized by means of Fourier-transform infrared spectroscopy and scanning electron microscopy. A proapoptotic drug, doxorubicin, was loaded into the hydrogels and the controlled release of the drug was assessed in the presence of α-amylase and ultimately it was evaluated to controlled-drug release in vitro and subsequently in killing cancer cells. Our results highlight the effectiveness of temporal drug delivery using α-amylase responsive hydrogels in killing cancer cells.


Subject(s)
Hydrogels/chemical synthesis , Starch/analogs & derivatives , alpha-Amylases/metabolism , Cell Death , Cell Line, Tumor , Cross-Linking Reagents/chemistry , Doxorubicin/chemistry , Doxorubicin/pharmacology , Humans , Spectroscopy, Fourier Transform Infrared , Starch/metabolism
13.
Pediatr Transplant ; 25(3): e13905, 2021 May.
Article in English | MEDLINE | ID: mdl-33179398

ABSTRACT

Bone marrow transplantation is the only curative treatment for beta-thalassemia major. Data on the co-transplantation of MSCs with HSCs in beta-thalassemia major patients are scarce. We aimed to investigate the outcomes of thalassemia major patients who underwent bone marrow-derived MSC co-transplantation with HSCs compared with those who only received HSCs. This prospective randomized study included patients with class III thalassemia major undergoing HSCT divided randomly into two groups: Thirty-three patients underwent co-transplantation of bone marrow-derived MSCs with HSCs, and 26 patients only received HSCs. Five-year OS, TFS, TRM, graft rejection rate, and GVHD were estimated. The 5-year OS was 66.54% (95% CI, 47.8% to 79.9%) in patients who underwent co-transplantation of MSCs with HSCs vs 76.92% (95% CI, 55.7% to 88.9%) in patients who only received HSCs (P = .54). No significant difference was observed in the 5-year TFS between the two groups (59.1% vs 69.2%; P = .49). The 5-year cumulative incidence of TRM was not statistically significant among patients who underwent co-transplantation of MSCs with HSCs (27.27%) vs those who only received HSCs (19.23%; P = .61). There was no statistically significant difference in graft rejection, acute GvHD, and chronic GvHD between the two groups. Based on our findings, the co-transplantation of MSCs and HSCs to class III thalassemia major patients does not alter their transplantation outcomes including OS, TFS, rejection rate, transplant-related mortality, and GvHD.


Subject(s)
Hematopoietic Stem Cell Transplantation , Mesenchymal Stem Cell Transplantation , beta-Thalassemia/therapy , Adolescent , Child , Cohort Studies , Combined Modality Therapy , Female , Follow-Up Studies , Humans , Male , Prospective Studies , Time Factors , Treatment Outcome , beta-Thalassemia/classification
14.
Article in English | MEDLINE | ID: mdl-32617114

ABSTRACT

Hepatocellular carcinoma (HCC) is the fifth most commonly diagnosed cancer and the second most common cause of cancer-related death worldwide. Sorafenib (Sora) is used as a targeted therapy for HCC treatment. Mesenchymal stem cells (MSCs) are applied as a new approach to fight malignancies. Drug resistance and side effects are the major concerns with Sora administration. The effect of using the combination of sorafenib and MSCs on tumor regression in xenograft HCC models was evaluated in this study. Methods and Materials. Human hepatocellular carcinoma cell lines (HepG2) were subcutaneously implanted into the flank of 18 nude mice. The animals were randomly divided into six groups (n = 3); each received Sora (oral), MSCs (IV injection), MSCs (local injection), Sora + MSCs (IV injection), Sora + MSCs (local injection), or no treatment (the control group). Six weeks after tumor implantation, the mice were scarified and tumoral tissues were resected in their entirety. Histopathological and immunohistochemical evaluations were used to measure tumor proliferation and angiogenesis. Apoptotic cells were quantified using the TUNEL assay. Results. No significant difference was found in the tumor grade among the treatment groups. Differentiation features of the tumoral cells were histopathologically insignificant in all the groups. Tumor necrosis was highest in the hpMSC (local) + Sora group. Tumor cell proliferation was reduced in hpMSC (local) + Sora-treated and hpMSC (IV) + Sora-treated mice compared with the other groups. Apoptotic-positive cells occupied a greater proportion in the Sora, hpMSC (IV) + Sora, and hpMSC (local) + Sora groups. Conclusion. A combination of chemotherapy and MSC can yield to more favorable results in the treatment of HCC.

15.
Int J Hematol Oncol Stem Cell Res ; 14(1): 11-18, 2020 Jan 01.
Article in English | MEDLINE | ID: mdl-32337010

ABSTRACT

Background: The present study investigated the patients with Chronic Myeloid Leukemia in chronic phase (CP-CML) who had been on the first- line Imatinib Mesylate (IM) therapy for a period of 84 months. Materials and Methods: This retrospective study was conducted in 295 newly-diagnosed CP-CML patients(age >18 years) who were admitted to the Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, Tehran during 1 January, 2009 to 30 December, 2016. Response to treatment was evaluated by molecular response assessment. Rates of IM dose adjustment, switching to another drug therapy, progression to Accelerate Phase (AP) and Blastic Crisis (BC) and long-term outcomes included Overall Survival (OS) and Progression Free Survival (PFS) were assesed. Results: Patients' average age was 41.7 years, and 52.9% were male. 44.4% of patients at the month 18 achieved Major Molecular Response (MMR). Progression to AP/BC occurred in 26 patients during 84 months, and the estimated rate of OS and PFS were 71.83 and 74.48, respectively. Among the patients who did not achieve MMR at month 18 , 61 patients were treated with IM ( 400 mg /day), and then after month 18, 24(39.3%) of whom achieved MMR. Dose adjustments occurred in 60 patients (20.33%). IM dose increase was observed in 53 patients who did not achive optimal response to imatinib or loss of optimal response. IM dose decrease was observed in 7 patients. 25 (8.47%) patients were switched to a different Tyrosine Kinase Inhibitor (TKI). Most of TKI changes(n=21) happened in patients who did not achieve optimal response to IM and TKI changes owing to adverse events of IM were observed in 4 patients.. Among the patients undergoing change in treatment, 24(43.75%) patients achieved MMR. Conclusion: Our data showed the high effectiveness of the change in the treatment of IM-resistant condition. Moreover, our finding suggests that imatinib be effective in Iranian patients after a long period of time compared to the referenced studies.

16.
Colloids Surf B Biointerfaces ; 190: 110884, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32113164

ABSTRACT

Montmorillonite (Mt) nanosheets are used in pharmaceutical products as both excipient and active ingredients. In addition, Mt can be used as a nanocarrier for oral delivery of drugs, including chemotherapy drugs, and as an embolic agent for tumor arterial embolization. It is noteworthy that, there is few conflicting evidence on the intrinsic antitumor activity of Mt. Hence, in this study, the antitumor potential of Mt was investigated using MRC-5, HT-29 and HepG2 cell lines. MTT assay revealed that, Mt possesses antiproliferative effect, which was concentration-dependent and affected by both protein level and cell type. However, this antiproliferative effect was not significantly affected by increasing the exposure time from 24 to 48 h. The results of flow-cytometry and qRT-PCR analyses showed that, Mt induced G0/G1-phase arrest in MRC-5 and HT-29 cells by modulating P21, P27 and Cyclin D1 genes, whereas it induced S-phase arrest in HepG2 cells probably by damaging DNA and up-regulating mTOR gene. The results also indicated that, Mt induced a high rate of apoptosis in all the cell lines by modulating anti/pro-apoptotic genes, as well as a rate of necrosis in HT-29. The apoptosis of MRC-5 and HT-29 cells was accompanied with up-regulation of P62 gene, suggesting autophagy-dependent apoptosis. In addition, in all the cell lines, Mt significantly enhanced the expression of executioner caspase-3. Based on these results, the biocompatible Mt nanosheets can act as antitumor agents. These findings may provide new applications of Mt in the field of cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Bentonite/pharmacology , Biocompatible Materials/pharmacology , Biological Products/pharmacology , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Bentonite/chemistry , Biocompatible Materials/chemistry , Biological Products/chemistry , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Drug Screening Assays, Antitumor , HT29 Cells , Hep G2 Cells , Humans , Particle Size , Surface Properties
17.
Drug Des Devel Ther ; 14: 185-194, 2020.
Article in English | MEDLINE | ID: mdl-32021103

ABSTRACT

INTRODUCTION: Acute myeloid leukemia (AML) is a type of blood disorder that exhibits uncontrolled growth and reduced ability to undergo apoptosis. Signal transducer and activator of transcription 3 (STAT3) is a family member of transcription factors which promotes carcinogenesis in most human cancers. This effect on AML is accomplished through deregulation of several critical genes, such as B cell lymphoma-extra-large (BCL-XL) which is anti-apoptotic protein. The aim of this study was to evaluate the effect of curcumin (CUR) and thalidomide (THAL) on apoptosis induction and also the alteration of the mRNA expression level of STAT3 and BCL-XL mRNA on AML cell line compounds. METHODS: The growth inhibitory effects of CUR and THAL and their combination were measured by MTT assay in U937 and KG-1 cell lines. The rates of apoptosis induction and cell cycle analysis were measured by concurrent staining with Annexin V and PI. The mRNA expression level of STAT3 and BCL-XL was evaluated by Real-Time PCR. RESULTS: CUR inhibited proliferation and induced apoptosis in both KG-1 and U937 cells and this effect increased by combination with THAL. The expression level of STAT3 and BCL-XL was significantly down-regulated in KG-1 cells after treatment by CUR and THAL and their combination. CONCLUSION: Overall, our findings suggested that down-regulation of STAT3 and BCL-XL mRNA expression in response to CUR and THAL treatment lead to inhibition of cell growth and induction of apoptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Curcumin/pharmacology , Leukemia, Myeloid, Acute/drug therapy , STAT3 Transcription Factor/antagonists & inhibitors , Thalidomide/pharmacology , bcl-X Protein/antagonists & inhibitors , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , STAT3 Transcription Factor/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured , bcl-X Protein/metabolism
18.
Cell J ; 22(1): 23-29, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31606963

ABSTRACT

OBJECTIVE: Multiple myeloma (MM) is an incurable plasma cell malignancy. Several genetic and epigenetic changes affect numerous critical genes expression status in this disorder. CDKN2A gene is expressed at low level in almost all cases with MM disease. The mechanism of this gene down-regulation has remained controversial. In the present study, we targeted EZH2 by microRNA-124 (miR-124) in L-363 cells and assessed following possible impact on CDKN2A gene expression and phenotypic changes. MATERIALS AND METHODS: In this experimental study, growth inhibitory effects of miR-124 were measured by MTT assay in L-363 cell line. Likewise, cell cycle assay was measured by flowcytometery. The expression levels of EZH2 and CDKN2A were evaluated by real-time quantitative reverse-transcription polymerase chain reaction (qRT-PCR). RESULTS: qRT-PCR results showed induction of EZH2 gene expression after transduction of cells with lentivector expressing miR-124. The expression of CDKN2A was also upregulated as the result of EZH2 supression. Coincide with gene expression changes, cell cycle analysis by flow-cytometry indicated slightly increased G1-arrest in miRtransduced cells (P<0.05). MTT assay results also showed a significant decrease in viability and proliferation of miRtransduced cells (P<0.05). CONCLUSION: It seems that assembling of H3K27me3 mark mediated by EZH2 is one of the key mechanisms of suppressing CDKN2A gene expression in MM disease. However, this suppressive function is applied by a multi-factor mechanism. In other words, targeting EZH2, as the core functional subunit of PRC2 complex, can increase expression of the downstream suppressive genes. Consequently, by increasing expression of tumor suppressor genes, myeloma cells are stopped from aberrant expansions and they become susceptible to regulated cellular death.

19.
Cell J ; 22(2): 193-202, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31721534

ABSTRACT

OBJECTIVE: Autophagy and apoptosis play key roles in cancer survival and pathogenesis and are governed by specific genes which have a dual role in both cell death and survival. Arsenic trioxide (ATO) and thalidomide (THAL) are used for treatment of many types of hematologic malignancies. ATO prevents the proliferation of cells and induces apoptosis in some cancer cells. Moreover, THAL has immunomodulatory and antiangiogenic effects in malignant cells. The aim of present study was to examine the effects of ATO and THAL on U937 and KG-1 cells, and evaluation of mRNA expression level of VEGFs genes, PI3K genes and some of autophagy genes. MATERIALS AND METHODS: In this in vitro experimental study, U937 and KG-1 cells were treated by ATO (0.4-5 µM) and THAL (5-100 µM) for 24, 48 and 72 hours. Cell viability was measured by MTT assay. The apoptosis rate and cell cycle arrest were evaluated by flow cytometry (Annexin/PI) and cell cycle flow cytometry analysis, respectively. The effect of ATO/THAL on mRNAs expression was evaluated by real-time polymerase chain reaction (PCR). RESULTS: ATO/THAL combination enhanced cell apoptosis in a dose-dependent manner. Also, ATO/THAL induced SubG1/ G1 phase arrest. mRNA expression levels of VEGFC (contrary to other VEGFs isoform), PI3K, AKT, mTOR, MEK1, PTEN, IL6, LC3 and P62 genes were upregulated in acute myeloid leukemia (AML) cells following treatment with ATO/THAL. CONCLUSION: Combined treatment with ATO and THAL can inhibit proliferation and invasion of AML cells by down-regulating ULK1 and BECLIN1 and up-regulating PTEN and IL6, and this effect was more marked than the effects of ATO and THAL alone.

20.
J Vasc Surg Venous Lymphat Disord ; 8(3): 445-451, 2020 05.
Article in English | MEDLINE | ID: mdl-31859244

ABSTRACT

OBJECTIVE: Lymphedema is a chronic debilitating disease characterized by the accumulation of fluid in the extremities as a result of lymphatic system impairment. Current treatments fail to restore the functionality and structural integrity of the lymphatic vessels lost in this condition. In this study, autologous mobilized peripheral blood stem cell transplantation was used and its potential efficacy and safety were evaluated in treating this condition. METHODS: Ten patients with primary lymphedema in the lower extremity received granulocyte-colony stimulating factor subcutaneously for 4 days, to stimulate stem cell mobilization, after which 200 to 250 mL of blood was drawn from each patient and used to collect stem cells. Mobilized stem cells were counted by flow cytometry with International Society of Hematotherapy and Graft Engineering method. In two sessions, 3 weeks apart, these stem cells were injected subcutaneously in the affected limb at approximately 80 points, along the lymphatic vessels. Each patient was followed for 6 months, during which changes in the limb volume and circumference were measured. Lymphangiogenesis was evaluated by biopsy, the lymphoscintigraphic transport index was calculated using Lymphoscintigraphy, and quality of life was surveyed. RESULTS: In this study, patients received on average 9.5 ± 6.8 × 108 mononuclear cells (which divided into 2 × 106 CD34+ cells for each session) in two sessions. The volume of the lower limbs decreased in 60% of patients. One patient showed a slight increase in the volume of lower limbs and three showed no change. The average limb volume was 4469.41 ± 1760.71 cm3, which on average differed from the average initial limb volume by 232.88 ± 392.53 cm3. Quality of life was reported as slightly increased in 60% of patients. The lymphoscintigraphic transport index suggested improvement in 60% of the patients. Likewise, tissue samples showed a 60% increase in lymphatic vessels. CONCLUSIONS: Subcutaneous injection of autologous hematopoietic stem cells harvested from peripheral blood into patients with primary lower limb lymphedema is feasible, potentially effective, and without serious adverse effects. However, a larger scale study with more patients is needed to validate our results. Last, to increase the effectiveness of this treatment, the optimal dose of cells injected and the requirement for additional growth factors need further study.


Subject(s)
Granulocyte Colony-Stimulating Factor/administration & dosage , Hematopoietic Stem Cell Mobilization , Lymphedema/surgery , Peripheral Blood Stem Cell Transplantation , Adult , Cell Separation , Female , Humans , Iran , Lower Extremity , Lymphedema/pathology , Lymphedema/physiopathology , Male , Middle Aged , Pilot Projects , Prospective Studies , Quality of Life , Recovery of Function , Time Factors , Transplantation, Autologous , Treatment Outcome , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...