Personalized Drug Combinations for the Treatment of Acute Myeloid Leukemia (AML) Patients

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3616-3616 ◽  
Author(s):  
Yunyi Kang ◽  
Trish Tran ◽  
Linda Zhang ◽  
Edward D. Ball ◽  
Carlo Piermarocchi ◽  
...  

Abstract Acute myeloid leukemia (AML) is a highly heterogeneous disorder characterized by the rapid clonal proliferation of blasts derived from hematopoietic progenitor cells, leading to failure of normal hematopoiesis. Although standard therapy, usually including idarubicin and cytarabine, has been used to achieve remission, the long-term survival rates remain low. To show that it is possible to improve existing AML standard therapy, we have used experimental algorithms we have recently developed (iterative algorithms that find effective combinations without a full combinatorial screening) to find selective drug combinations for AML cells. The first search was done using the AML cell line KG-1 for the initial steps and cells from an AML patient for the final step. A normal fibroblast cell line (IMR-90) and normal blood progenitor cells were used as controls for toxicity. Drugs were chosen from those already approved or in clinical trials for AML. As shown in Figure 1, this experimental search was able to achieve selective killing of AML patient cells with a ratio close to 100:1 with respect to non-cancer control cells. To refine the choice of drugs to be used in the search, we recently developed a network model of AML intracellular signal transduction (see www.leukemianetworks.org). The present version (AML 2.1) includes 5667 proteins and 22,218 interactions. The network can be used to integrate genomic and gene expression information from individual patients and to provide a shortlist of drugs to be tested further. Using literature data and our network model we identified three compounds as promising for AML drug combinations: the MEK inhibitor PD0325901, the FLT3 inhibitor Quizartinib and the CDK 4/6 inhibitor Palbociclib. For this experiment, normal progenitor blood cells were the controls. Figure 2 shows the results of 64 experimental tests of the response to different combinations of these drugs. The two AML patients clearly exhibited different drug sensitivities, further supporting the necessity of personalized drug combinations. To improve the clinical relevance of these in vitro studies, we investigated the correlation between clinical and in vitro response to standard AML therapy, using a set of media containing 16 cytokines/growth factors in different combinations. These molecules have been selected using three methods: ligands acting on receptors that are part of gene signatures with prognostic significance in AML; cytokines that promote hematopoietic differentiation of stem cells; and ligands of receptors identified as playing an important role in AML in a recent RNAi screen. During several iterations of our combinatorial search algorithm we studied 185 different media. Figure 3 shows the correlation between clinical response (residual blasts after 28 days of therapy) and response to the same therapy in vitro after 72 h achieved in the last iteration of the search, in a study of six patients. The medium with the highest correlation had a correlation coefficient of 0.99 (p=0.0002). The in vitro response in this medium is clearly able to distinguish patients that respond to standard therapy vs those that do not, as can be seen from the inset of Fig. 3. Some cytokines (BMP-4 and IL-4) were always absent in the group of 14 media with lower correlation shown on the right of Figure 3. The remarkable variation in correlation with clinical response of these media, spanning correlation coefficient values from 0.99 to 0.004, clearly show the importance of optimizing the in vitro microenvironment for prediction of patient drug response. In summary, we identified drug combinations that selectively kill AML primary cells and we optimized media that improve the in vitro prediction of clinical drug response. These methods can together assist in the personalization of AML therapy using patientÕs cells. Figure 1. Selective drug combinations for AML primary cells. Selectivity is the ratio between AML and control cell viability. Figure 1. Selective drug combinations for AML primary cells. Selectivity is the ratio between AML and control cell viability. Figure 2. Selective drug combinations for AML primary cells from two patients, using multiple doses and combinations of Quizartinib, PD-0325901 and Palbociclib. Patient 2005-9 responded to MEK inhibitor PD-0325901 whereas Patient 2010-7 responded to FLT3 inhibitor Quizartinib. Figure 2. Selective drug combinations for AML primary cells from two patients, using multiple doses and combinations of Quizartinib, PD-0325901 and Palbociclib. Patient 2005-9 responded to MEK inhibitor PD-0325901 whereas Patient 2010-7 responded to FLT3 inhibitor Quizartinib. Figure 3. A pilot study showing the clinically predictive power of optimized media. The inset shows the best medium. Please note the strong effect of media composition on clinical correlation. Figure 3. A pilot study showing the clinically predictive power of optimized media. The inset shows the best medium. Please note the strong effect of media composition on clinical correlation. Disclosures Piermarocchi: Salgomed Inc.: Equity Ownership. Paternostro:Salgomed Inc.: Equity Ownership.

2021 ◽  
Vol 2 ◽  
Author(s):  
Selma Ben Hamouda ◽  
Maria Angélica Miglino ◽  
Gustavo de Sá Schiavo Matias ◽  
Guy Beauchamp ◽  
Jean-Pierre Lavoie

Asthma is associated with increased deposition and altered phenotype of airway smooth muscle (ASM) cells. However, little is known about the processes responsible for these changes. It has been suggested that alterations of the extracellular matrix (ECM) contribute to the remodeling of ASM cells in asthma. Three-dimensional matrices allow the in vitro study of complex cellular responses to different stimuli in a close-to-natural environment. Thus, we investigated the ultrastructural and genic variations of ASM cells cultured on acellular asthmatic and control bronchial matrices. We studied horses, as they spontaneously develop a human asthma-like condition (heaves) with similarities to chronic pulmonary changes observed in human asthma. Primary bronchial ASM cells from asthmatic (n = 3) and control (n = 3) horses were cultured on decellularized bronchi from control (n = 3) and asthmatic (n = 3) horses. Each cell lineage was used to recellularize six different bronchi for 41 days. Histomorphometry on HEPS-stained-recellularized matrices revealed an increased ASM cell number in the control cell/control matrix (p = 0.02) and asthmatic cell/control matrix group (p = 0.04) compared with the asthmatic cell/asthmatic matrix group. Scan electron microscopy revealed a cell invasion of the ECM. While ASM cells showed high adhesion and proliferation processes on the control ECM, the presence of senescent cells and cellular debris in the asthmatic ECM with control or asthmatic ASM cells suggested cell death. When comparing asthmatic with control cell/matrix combinations by targeted next generation sequencing, only AGC1 (p = 0.04), MYO10 (p = 0.009), JAM3 (p = 0.02), and TAGLN (p = 0.001) were differentially expressed out of a 70-gene pool previously associated with smooth muscle remodeling. To our knowledge, this is the first attempt to evaluate the effects of asthmatic ECM on an ASM cell phenotype using a biological bronchial matrix. Our results indicate that bronchial ECM health status contributes to ASM cell gene expression and, possibly, its survival.


1998 ◽  
Vol 141 (3) ◽  
pp. 647-662 ◽  
Author(s):  
David A. Potter ◽  
Jennifer S. Tirnauer ◽  
Richard Janssen ◽  
Dorothy E. Croall ◽  
Christina N. Hughes ◽  
...  

Previous studies suggest that the Ca2+-dependent proteases, calpains, participate in remodeling of the actin cytoskeleton during wound healing and are active during cell migration. To directly test the role that calpains play in cell spreading, several NIH-3T3– derived clonal cell lines were isolated that overexpress the biological inhibitor of calpains, calpastatin. These cells stably overexpress calpastatin two- to eightfold relative to controls and differ from both parental and control cell lines in morphology, spreading, cytoskeletal structure, and biochemical characteristics. Morphologic characteristics of the mutant cells include failure to extend lamellipodia, as well as abnormal filopodia, extensions, and retractions. Whereas wild-type cells extend lamellae within 30 min after plating, all of the calpastatin-overexpressing cell lines fail to spread and assemble actin-rich processes. The cells genetically altered to overexpress calpastatin display decreased calpain activity as measured in situ or in vitro. The ERM protein ezrin, but not radixin or moesin, is markedly increased due to calpain inhibition. To confirm that inhibition of calpain activity is related to the defect in spreading, pharmacological inhibitors of calpain were also analyzed. The cell permeant inhibitors calpeptin and MDL 28, 170 cause immediate inhibition of spreading. Failure of the intimately related processes of filopodia formation and lamellar extension indicate that calpain is intimately involved in actin remodeling and cell spreading.


2021 ◽  
Vol 22 (18) ◽  
pp. 9986
Author(s):  
Giulia Brunello ◽  
Kathrin Becker ◽  
Luisa Scotti ◽  
Dieter Drescher ◽  
Jürgen Becker ◽  
...  

Several decontamination methods for removing biofilm from implant surfaces during surgical peri-implantitis treatment have been reported, including the intraoperative usage of chlorhexidine (CHX)-based antiseptics. There is a lack of information on possible adverse effects on bone healing. The study aimed to examine the impact of three CHX-based mouthwashes on osteoblast-like cells (SaOS-2) in vitro. Cells were cultured for three days in 96-well binding plates. Each well was randomly treated for either 30, 60 or 120 s with 0.05% CHX combined with 0.05% cetylpyridinium chloride (CPC), 0.1% CHX, 0.2% CHX or sterile saline (NaCl) as control. Cell viability, cytotoxicity and apoptosis were assessed at day 0, 3 and 6. Cell viability resulted in being higher in the control group at all time points. At day 0, the CHX 0.2 group showed significantly higher cytotoxicity values compared to CHX 0.1 (30 s), CHX + CPC (30 s, 60 s and 120 s) and control (60 s and 120 s), while no significant differences were identified between CHX + CPC and both CHX 0.1 and NaCl groups. All test mouthwashes were found to induce apoptosis to a lower extent compared to control. Results indicate that 0.2% CHX presented the highest cytotoxic effect. Therefore, its intraoperative use should be carefully considered.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4931-4931
Author(s):  
Monica Hellesøy ◽  
Katarzyna Wnuk-Lipinska ◽  
Anna Boniecka ◽  
Eline Milde Nævdal ◽  
Hakon Reikvam ◽  
...  

Abstract Axl is a member of the Tyro3, Axl, Mer (TAM) receptor tyrosine kinase family that regulate a wide range of cellular functions, including cell survival, proliferation, migration/invasion and adhesion. Axl has been shown to play a key role in the survival and metastasis of many tumors, and has also been found to be upregulated and constitutively active in human AML. Indeed, Axl has been reported as an independent prognostic marker and a potential novel therapeutic target in AML. BGB324 is a first-in-class highly selective small molecule inhibitor of Axl. BGB324 has been shown to be safe and well tolerated in clinical safety studies in healthy volunteers at doses up to 1500 mg/day with a predictable PK profile and long plasma half-life, and is currently in phase I b clinical trials for AML and non-small cell lung cancer. In this study, we use phosphoflow cytometry to measure changes in signal transduction nodes in single AML cells treated with BGB324. We are applying this approach to monitor signaling profiles in primary AML cells harvested from patients undergoing BGB324 treatment. Results: The human AML cell line MOLM13 was treated in vitro with BGB324 (0.5 and 1µM for 1 hour) and analyzed for signal transduction changes by phosphoflow cytometry. We found a significant reduction in phosphorylation of Axl (pY779), Akt(pS473), Erk1/2(pT202/Y204) and PLCɣ1(pY783). Next we established a systemic MOLM13 preclinical AML model in NOD/SCID mice. The mice were treated with 25 or 50 mg/kg BGB324 until moribund (up to 16 days). We found a dose-dependent and significant increase in overall survival in BGB324-treated mice. We further investigated intracellular signaling in BGB324-treated cells in vivo. Mice carrying systemic AML disease (MOLM13) were treated with BGB324 at 50mg/kg for 4 days, and we monitored CD33/45-positive MOLM13 cells harvested from spleen and bone marrow by flow cytometry. BGB324-treated mice showed a significant reduction in pErk and pPLCɣ1 relative to mice in the control group. PBMCs from peripheral blood of AML patients treated with BGB324 400 mg x1 at day 1 and 2, and thereafter 100 mg daily were collected for single cell signal profiling of signal transduction changes by conventional flow cytometry (phospho-flow) and mass cytometry (CyTOF). Preliminary phopho-flow analyses show decrease of pAkt(T308) and pPLCgamma1(Y783) in one patient. Further analyses are ongoing and will be presented. Figure 1. In vitro response to 1 hour BGB324 treatment in human AML cell line MOLM13 at 0.5 and 1µM doses. Response was evaluated in pAxl, pErk1/2, pAkt and pPLCγ1. n=3, *p≤0.05, **p≤0.005. Figure 1. In vitro response to 1 hour BGB324 treatment in human AML cell line MOLM13 at 0.5 and 1µM doses. Response was evaluated in pAxl, pErk1/2, pAkt and pPLCγ1. n=3, *p≤0.05, **p≤0.005. Figure 2. Dose-dependent response in overall survival in a MOLM13 systemic xenograft model (n=10). Figure 2. Dose-dependent response in overall survival in a MOLM13 systemic xenograft model (n=10). Figure 3. Response to BGB324-treatment in pErk, pPLCγ1 and pAkt in CD33/CD45-positive cells harvested from spleens (left) and bone marrows (right) of mice with systemic MOLM13 xenografts. n=5, *p≤0.05, **p≤0.005. Figure 3. Response to BGB324-treatment in pErk, pPLCγ1 and pAkt in CD33/CD45-positive cells harvested from spleens (left) and bone marrows (right) of mice with systemic MOLM13 xenografts. n=5, *p≤0.05, **p≤0.005. Disclosures Hellesøy: BerGenBio AS: Other: Previous employee. Stock option holder. Wnuk-Lipinska:BerGenBio AS: Employment. Boniecka:BerGenBio AS: Employment. Nævdal:BerGenBio AS: Employment. Loges:BerGenBio: Honoraria, Other: travel support, Research Funding. Cortes:Teva: Research Funding; BMS: Consultancy, Research Funding; Novartis: Consultancy, Research Funding; Pfizer: Consultancy, Research Funding; BerGenBio AS: Research Funding; Ariad: Consultancy, Research Funding; Astellas: Consultancy, Research Funding; Ambit: Consultancy, Research Funding; Arog: Research Funding; Celator: Research Funding; Jenssen: Consultancy. Lorens:BerGenBio AS: Employment, Equity Ownership. Micklem:BerGenBio AS: Employment, Equity Ownership. Gausdal:BerGenBio AS: Employment. Gjertsen:Haukeland University Hospital: Research Funding.


2021 ◽  
Vol 22 (15) ◽  
pp. 8132
Author(s):  
Jennifer Zhang ◽  
Oscar Hou-In Chou ◽  
Yiu-Lam Tse ◽  
Kwong-Man Ng ◽  
Hung-Fat Tse

Inherited cardiomyopathies are among the major causes of heart failure and associated with significant mortality and morbidity. Currently, over 70 genes have been linked to the etiology of various forms of cardiomyopathy, some of which are X-linked. Due to the lack of appropriate cell and animal models, it has been difficult to model these X-linked cardiomyopathies. With the advancement of induced pluripotent stem cell (iPSC) technology, the ability to generate iPSC lines from patients with X-linked cardiomyopathy has facilitated in vitro modelling and drug testing for the condition. Nonetheless, due to the mosaicism of the X-chromosome inactivation, disease phenotypes of X-linked cardiomyopathy in heterozygous females are also usually more heterogeneous, with a broad spectrum of presentation. Recent advancements in iPSC procedures have enabled the isolation of cells with different lyonisation to generate isogenic disease and control cell lines. In this review, we will summarise the current strategies and examples of using an iPSC-based model to study different types of X-linked cardiomyopathy. The potential application of isogenic iPSC lines derived from a female patient with heterozygous Danon disease and drug screening will be demonstrated by our preliminary data. The limitations of an iPSC-derived cardiomyocyte-based platform will also be addressed.


2020 ◽  
Vol 10 (1) ◽  
Author(s):  
Evelina Folkesson ◽  
Barbara Niederdorfer ◽  
Vu To Nakstad ◽  
Liv Thommesen ◽  
Geir Klinkenberg ◽  
...  

AbstractDrug combinations have been proposed to combat drug resistance, but putative treatments are challenged by low bench-to-bed translational efficiency. To explore the effect of cell culture format and readout methods on identification of synergistic drug combinations in vitro, we studied response to 21 clinically relevant drug combinations in standard planar (2D) layouts and physiologically more relevant spheroid (3D) cultures of HCT-116, HT-29 and SW-620 cells. By assessing changes in viability, confluency and spheroid size, we were able to identify readout- and culture format-independent synergies, as well as synergies specific to either culture format or readout method. In particular, we found that spheroids, compared to 2D cultures, were generally both more sensitive and showed greater synergistic response to combinations involving a MEK inhibitor. These results further shed light on the importance of including more complex culture models in order to increase the efficiency of drug discovery pipelines.


2007 ◽  
Vol 361-363 ◽  
pp. 1001-1004 ◽  
Author(s):  
Barbara Bracci ◽  
Milena Fini ◽  
Silvia Panzavolta ◽  
Paola Torricelli ◽  
Adriana Bigi

We recently developed a new biomimetic calcium phosphate bone cement enriched with gelatin (GEL-CP) which exhibits improved mechanical properties with respect to the control cement (C-CP) and a good response to osteoblast-like cells. In this work, we have extended the investigation to primary culture of osteoblasts derived from normal (N-OB) and osteopenic (O-OB) sheep bones cultured on samples of GEL-CP, and their behavior was compared to that of cells cultured on C-CP as control. Cell morphology, proliferation, and differentiation were evaluated at 3 and 7 days. Preliminary in vivo tests were carried out onto critical size defects in the radius diaphysis of rats.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3705-3705
Author(s):  
Wenqing Wang ◽  
Avni Awani ◽  
Andrew Devilbiss ◽  
Thomas Mathews ◽  
Daniel Thomas ◽  
...  

While hematopoietic stem and progenitor cells (HSPCs) were thought to rely mainly on glycolysis for energy supply, emerging evidence suggests that defects in mitochondrial functions can impact HSPC development with respect to self-renewal, differentiation and aging. The exact mechanisms underlying metabolic reprogramming and cell fate decisions during human hematopoiesis, however, remain elusive. Biallelic mutations in the mitochondrial enzyme adenylate kinase 2 (AK2), cause reticular dysgenesis (RD), one of the most profound forms of severe combined immunodeficiency (SCID). AK2 catalyzes the interconversion between adenine nucleotides and thereby controls the availability of ADP for oxidative phosphorylation. Clinically, RD patients not only present with profound lymphopenia, typical for classic SCID, but also suffer from severe congenital neutropenia. The developmental arrest across the T, NK and granulocytic lineages suggests that AK2 deficiency causes a metabolic defect with global impact on hematopoiesis. Our prior work in induced pluripotent stem cells (iPSCs) from RD patients has shown that maturation-arrested iPSC-derived HSPCs exhibit increased oxidative stress and an energy-depleted adenine nucleotide profile, suggesting that AK2-regulated mitochondrial bioenergetics play an integral role in HSPC differentiation. Therefore, RD serves as an excellent model to study the impact of mitochondrial metabolism during human HSPC development. Methods: Since iPSCs do not recapitulate definitive hematopoiesis, we developed an AK2 biallelic knock-out model in primary human HSPCs using CRISPR/Cas9 gene editing. Employing a homologous recombination-mediated dual color reporter strategy, we were able to select for HSPCs with biallelic AK2 knock-out. HSPCs edited at the safe harbor AAVS1 site were used as a control. FACS purified AK2-/- and AAVS1-/- HSPCs were in vitro differentiated along the granulocytic lineage, and cells at various differentiation stages were sorted for RNA-seq and metabolomics analysis. Results: We analyzed the myeloid differentiation potential of AK2-/- HSPCs in vitro. Compared to AAVS1-/- controls, AK2-/- HSPCs displayed a severely decreased colony forming potential of both myeloid and erythroid lineages. In addition, AK2-/- HSPCs showed a granulocytic maturation arrest at the HLA-DR-, CD117+ promyelocyte stage, consistent with the characteristic phenotype observed in RD patients. We then performed RNA-seq studies on in vitro differentiated promyelocyte and neutrophil subpopulations derived from AK2-/- and control HSPCs. The RNA-seq analysis showed differential gene expression in glutathione metabolism and IL-10 signaling pathways, suggesting an increase in oxidative stress and inflammation, respectively, caused by AK2 deficiency. In addition, genes implicated in antimicrobial function and granule synthesis were downregulated in AK2-/- neutrophils, suggesting a functional defect. Liquid chromatography-mass spectrometry (LC-MS/MS) studies to delineate differences in metabolite profile conferred by AK2 deficiency at different stages of HSPC development are currently in progress. Conclusions: We have established the first cell-traceable biallelic AK2 CRISPR knock-out model in primary human HSPCs that recapitulates the myeloid phenotype of RD patients. This model allows us to profile AK2 knock-out cells at different developmental stages. AK2-/- granulocyte precursors showed a transcriptional signature suggestive of worsening oxidative stress, inflammation and defective effector cell functions during maturation. To understand the mechanistic underpinnings for these observations we are now using a global metabolomics approach to profile the changes in energy metabolites that occur during development in AK2-deficient and control HSPC subpopulations. Understanding how metabolism governs differentiation and self-renewal of human HSPCs has important translational implications to improve hematopoietic stem cell products and transplantation outcomes. Disclosures Morrison: Frequency Therapeutics: Consultancy, Membership on an entity's Board of Directors or advisory committees; OncoMed Pharmaceuticals: Equity Ownership; GI Therapeutics: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Kolon Gene Therapeutics: Consultancy; Protein Fluidics: Other: Stock Options.


2019 ◽  
Vol 6 (2) ◽  
pp. 48
Author(s):  
Lutfi Chabib ◽  
Farida Hayati ◽  
Rizki Awaluddin ◽  
Muh Iqbal Pangestu

ABSTRAK Kangkung darat (Ipomea reptans Poir) adalah salah satu tanaman di Indonesia yang secara empiris telah dimanfaatkan oleh masyarakat sebagai obat untuk terapi Diabetes Melitus (DM).  Riset sebelumnya telah dilakukan untuk memperoleh data ilmiah dari pemanfaatan kangkung darat sebagai terapi DM, dan pengembangannya dalam bentuk sediaan Self Nano Emulsifying Drug Delivery System (SNEDDS). Penelitian ini bertujuan untuk mengetahui sitotoksisitas sediaan SNEDDS ekstrak daun kangkung terstandar dengan menggunakan metode MTT (Methyl-Thiazolyl-Tetrazolium). Pengujian MTT dilakukan dengan cara well plate diplotkan terlebih dahulu untuk sampel uji 70 sumuran (well), kontrol sel 6 sumuran, dan kontrol media 6 sumuran.  Terakhir tiap sumuran ditambahkan stopper SDS sebanyak 100µl. Plate dibungkus alumunium foil dan diinkubasi semalaman pada suhu ruang. Hari berikutnya dilakukan pembacaan absorbansi sampel, kontrol media, dan kontrol sel menggunakan ELISA reader. Uji sitotoksik SNEEDS kangkung pada sel vero diperoleh hasil bahwa SNEEDS kangkung tidak menyebabkan kematian pada sel uji. Pada pengujian sel dilakukan pemaparan sampel excipient dari SNEEDS kangkung sebagai pembanding, dengan hasil menyebabkan kematian <50% sel uji pada dua kadar tertinggi dari sampel excipient. Namun pada hasil uji SNEEDS kangkung diperoleh data bahwa toksisitas excipient pada kadar tersebut tidak mempengaruhi timbulnya toksisitas pada sampel SNEEDS kangkung. Sehingga berdasarkan uji in vitro, SNEEDS kangkung tidak toksik terhadap sel vero. Kata kunci: Ipomoea reptans, sitotoksisitas, SNEDDS  ABSTRACT Kangkong (Ipomea reptans Poir) is a type of Indonesian plant empirically utilized by many people to treat diabetes mellitus (DM). A study has been conducted to obtain scientific data from the use of kangkong for DM treatment as well as to develop kangkong in SNEDDS preparation. This current study aimed to examine the cytotoxicity of kangkong leaf standardized extract in SNEDDS preparation through the MTT (Methyl-Thiazolyl-Tetrazolium) method. MTT assay was performed by initially plotting well plates for test sample (70 wells), control cell (6 wells), and control medium (6 wells). As much as 100µl SDS stopper was added into each well, and then plates were wrapped in aluminum foil and incubated all night at ambient temperature. On the following day, the absorbance of test sample, control medium, and control cell was identified using ELISA reader. The cytotoxicity test of kangkong SNEDDS on Vero cell lines showed that kangkong SNEDDS did not cause cell death. The cell was tested through exposure of excipient sample from kangkong SNEDDS as a comparison, resulting in <50% cell death by the two highest concentrations of excipient sample. However, the test result of kangkong SNEDDS indicated that excipient toxicity at such concentrations did not affect kangkong SNEDDS. Therefore, based on in vitro test, kangkong SNEDDS is not toxic against Vero cell lines. Keywords: Ipomoea reptans, cytotoxicity, SNEDDS


Sign in / Sign up

Export Citation Format

Share Document