Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Pediatr Blood Cancer ; 71(3): e30813, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38110844

ABSTRACT

BACKGROUND: 6-Mercaptopurine (6MP) is the mainstay chemotherapy for acute lymphoblastic leukemia (ALL) and is conventionally available as 50 mg tablets. A new 6MP powder for oral suspension (PFOS 10 mg/mL) was developed recently by IDRS Labs, India, intended for pediatric use. A comparative pharmacokinetics of PFOS with T. mercaptopurine was conducted to determine the dose equivalence. METHODS: An open-label, randomized, two-treatment, two-period, two-sequence, single oral dose, crossover, bioequivalence study was conducted on 51 healthy adult subjects. Post hoc, a population pharmacokinetic (PopPK) model was developed using the healthy volunteer data to perform simulations with various PFOS doses and select a bioequivalent dose. Further, to confirm the safety of PFOS in pediatrics, a simulation of 6MP and 6-thioguanine exposures was performed by incorporating the formulation-specific parameters derived from the healthy volunteer study into the PopPK model in childhood ALL available in literature. RESULTS: The 6MP PFOS had 47% higher oral bioavailability compared to the reference product. Simulations using a two-compartmental PopPK model with dissolution and transit compartments showed that 40 mg of PFOS was found to be equivalent to 50 mg tablets. The simulated 6-thioguanine nucleotide concentrations in children using the dose adjusted for PFOS were between 114 and 703.6 pmol/8 × 108 RBC, which was within the range reported in pediatric ALL studies. CONCLUSION: 6MP PFOS 10 mg/mL should be administered at a 20% lower dose than the tablet to achieve comparable exposure. 6MP PFOS addresses an unmet medical need for a liquid formulation of 6MP in the Indian subcontinent.


Subject(s)
Mercaptopurine , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Adult , Child , Humans , Administration, Oral , Cross-Over Studies , Mercaptopurine/administration & dosage , Powders , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Tablets , Therapeutic Equivalency , Thioguanine
2.
Acta Pharmacol Sin ; 45(7): 1506-1519, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38480835

ABSTRACT

Combining radiotherapy with Nrf-2 inhibitor holds promise as a potential therapeutic strategy for radioresistant lung cancer. Here, the radiosensitizing efficacy of a synthetic glucocorticoid clobetasol propionate (CP) in A549 human lung cancer cells was evaluated. CP exhibited potent radiosensitization in lung cancer cells via inhibition of Nrf-2 pathway, leading to elevation of oxidative stress. Transcriptomic studies revealed significant modulation of pathways related to ferroptosis, fatty acid and glutathione metabolism. Consistent with these findings, CP treatment followed by radiation exposure showed characteristic features of ferroptosis in terms of mitochondrial swelling, rupture and loss of cristae. Ferroptosis is a form of regulated cell death triggered by iron-dependent ROS accumulation and lipid peroxidation. In combination with radiation, CP showed enhanced iron release, mitochondrial ROS, and lipid peroxidation, indicating ferroptosis induction. Further, iron chelation, inhibition of lipid peroxidation or scavenging mitochondrial ROS prevented CP-mediated radiosensitization. Nrf-2 negatively regulates ferroptosis through upregulation of antioxidant defense and iron homeostasis. Interestingly, Nrf-2 overexpressing A549 cells were refractory to CP-mediated ferroptosis induction and radiosensitization. Thus, this study identified anti-psoriatic drug clobetasol propionate can be repurposed as a promising radiosensitizer for Keap-1 mutant lung cancers.


Subject(s)
Clobetasol , Ferroptosis , Lung Neoplasms , Mitochondria , NF-E2-Related Factor 2 , Reactive Oxygen Species , Humans , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/antagonists & inhibitors , Ferroptosis/drug effects , Reactive Oxygen Species/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/radiation effects , Clobetasol/pharmacology , Radiation-Sensitizing Agents/pharmacology , A549 Cells , Lipid Peroxidation/drug effects , Oxidative Stress/drug effects
3.
Phytother Res ; 38(3): 1555-1573, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38281735

ABSTRACT

Anti-inflammatory and immune suppressive agents are required to moderate hyper-activation of lymphocytes under disease conditions or organ transplantation. However, selective disruption of mitochondrial redox has not been evaluated as a therapeutic strategy for suppression of T-cell-mediated pathologies. Using mitochondrial targeted curcumin (MitoC), we studied the effect of mitochondrial redox modulation on T-cell responses by flow cytometry, transmission electron microscopy, transcriptomics, and proteomics, and the role of Nrf2 was studied using Nrf2- /- mice. MitoC decreased mitochondrial TrxR activity, enhanced mitochondrial ROS (mROS) production, depleted mitochondrial glutathione, and suppressed activation-induced increase in mitochondrial biomass. This led to suppression of T-cell responses and metabolic reprogramming towards Treg differentiation. MitoC induced nuclear translocation and DNA binding of Nrf2, leading to upregulation of Nrf2-dependent genes and proteins. MitoC-mediated changes in mitochondrial redox and modulation of T-cell responses are abolished in Nrf2- /- mice. Restoration of mitochondrial thiols abrogated inhibition of T-cell responses. MitoC suppressed alloantigen-induced lymphoblast formation, inflammatory cytokines, morbidity, and mortality in acute graft-versus-host disease mice. Disruption of mitochondrial thiols but not mROS increase inculcates an Nrf2-dependent immune-suppressive disposition in T cells for the propitious treatment of graft-versus-host disease.


Subject(s)
Curcumin , Curcumin/analogs & derivatives , Graft vs Host Disease , Animals , Mice , Curcumin/pharmacology , NF-E2-Related Factor 2/metabolism , T-Lymphocytes , Disease Models, Animal , Graft vs Host Disease/metabolism , Graft vs Host Disease/pathology , Sulfhydryl Compounds/metabolism , Sulfhydryl Compounds/pharmacology
4.
Br J Haematol ; 200(4): 429-439, 2023 02.
Article in English | MEDLINE | ID: mdl-36323643

ABSTRACT

Management of acquired immunodeficiency syndrome (AIDS)-related diffuse large B-cell (DLBCL) and plasmablastic lymphomas (PBL) poses significant challenges. The evidence supports use of dose-adjusted EPOCH (etoposide, prednisone, vincristine, cyclophosphamide and doxorubicin) with or without rituximab as first-line therapy. The need for central venous access, growth factors and significant toxicities limits its use in resource-constrained settings. To address these challenges, we have developed a novel regimen, CVEP (cyclophosphamide, vinblastine, etoposide, and prednisolone) based on the pharmacodynamic principles of dose-adjusted EPOCH. This single-centre phase II study evaluated the efficacy and safety of CVEP regimen in patients with de novo systemic AIDS-related DLBCL and PBL. The primary objective was complete response (CR) rates as assessed by positron emission tomography-computed tomography. The secondary objectives were incidence of Grade 3/4 toxicities, toxicities requiring hospitalisation, and disease-free survival. From May 2011 to February 2017, 42 patients were enrolled. At the end of therapy the CR rates were 69% (29/42) in the intention-to-treat population and 80.5% (29/36) in evaluable patients. At a median follow-up of 69 months, the 5-year disease-free survival was 65.3%. Out of 217 cycles administered, febrile neutropenia occurred in 19.3% and hospitalisation was required in 18.3% of cycles. There were two treatment-related deaths. The CVEP regimen is an active and safe regimen for AIDS-related DLBCL and PBL.


Subject(s)
Acquired Immunodeficiency Syndrome , Lymphoma, Large B-Cell, Diffuse , Humans , Etoposide/adverse effects , Vinblastine/adverse effects , Acquired Immunodeficiency Syndrome/drug therapy , Prednisolone/adverse effects , Cyclophosphamide/adverse effects , Prednisone/therapeutic use , Vincristine/adverse effects , Doxorubicin/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects
5.
IUBMB Life ; 75(10): 811-829, 2023 10.
Article in English | MEDLINE | ID: mdl-37072689

ABSTRACT

Aim of the present study was to test whether ionizing radiation (IR) treatment along with 3,3'-diselenodipropionic acid (DSePA), a redox active organodiselenide achieved better tumor control by suppressing the growth and migration of lung cancer cells. The results indicated that post-IR (2 Gy) treatment of DSePA (5 µM) led to a significantly higher cell death as compared to that of DSePA and IR treatments separately. Importantly, combinatorial treatment also showed reduction in the proportion of cancer stem cells and the clonogenic survival of A549 cells. The mechanistic studies indicated that combinatorial treatment although exhibited reductive environment (marked by decrease in ROS and increase of GSH/GSSG) at early time points (2-6 h postradiation), slowed DNA repair, inhibited epithelial-mesenchymal transition (EMT)/cell migration and induced significant level of apoptosis. DSePA mediated suppression of ATM/DNAPKs/p53 (DNA damage response signaling) and Akt/G-CSF (EMT) pathways appeared to be the major mechanism responsible for its radio-modulating activity. Finally, the combined treatment of IR (2 Gy × 4) and DSePA (0.1-0.25 mg/kg body weight daily through oral gavage) showed a significantly higher tumor suppression of the A549 xenograft as compared to that of DSePA and IR treatments separately in the mouse model. In conclusion, post-IR treatment of DSePA augmented cell kill by inhibiting DNA repair and cell migration in A549 cells.


Subject(s)
Apoptosis , Lung Neoplasms , Mice , Animals , Humans , A549 Cells , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/radiotherapy , DNA Repair , Cell Movement , Cell Line, Tumor
6.
Br J Clin Pharmacol ; 89(11): 3247-3261, 2023 11.
Article in English | MEDLINE | ID: mdl-37259249

ABSTRACT

Over the last three to four decades, Therapeutic Drug Monitoring (TDM) has shaped itself as therapeutic drug management, an integral component of precision medicine. The practice of TDM is not extensive in India, despite being one of the fastest-growing economies in the world. It is currently limited to a few academic medical centres and teaching hospitals. Apart from the immunosuppressive drugs, several other therapeutic areas, such as anticancer, antifungal, antibiotic and antitubercular, have demonstrated great potential to improve patient outcomes in Indian settings. Factors such as the higher prevalence of nutritional deficiencies, tropical diseases, widespread use of alternative medicines, unalike pharmacogenomics and sparse population-specific data available on therapeutic ranges of several drugs make the population of this subcontinent unique regarding the relevance of TDM. Despite the impact of TDM in clinical science and its widespread application, TDM has failed to receive the attention it deserves in India. This review intends to bring out a strength, weakness, opportunity and threats (SWOT) analysis for TDM in India so that appropriate steps for fostering the growth of TDM could be envisioned. The need of the hour is the creation of a cooperative group including all the stakeholders, such as TDM professionals, clinicians and the government and devising a National Action Plan to strengthen TDM. Nodal TDM centres should be established, and pilot programmes should be rolled out to identify the thrust areas for TDM in the country, capacity building and creating awareness to integrate TDM into mainstream clinical medicine.


Subject(s)
Drug Monitoring , Immunosuppressive Agents , Humans , Antitubercular Agents/therapeutic use , India
7.
Oncologist ; 27(3): 165-e222, 2022 03 11.
Article in English | MEDLINE | ID: mdl-35274724

ABSTRACT

BACKGROUND: This phase I dose de-escalation study aimed to assess the tolerability, safety, pharmacokinetics (PK), and efficacy of sequentially decreasing doses of sorafenib in combination (SAM) with atorvastatin (A, 10 mg) and metformin (M, 500 mg BD) in patients with advanced hepatocellular carcinoma (HCC). METHODS: Patients were enrolled in 1 of 4 sequential cohorts (10 patients each) of sorafenib doses (800 mg, 600 mg. 400 mg, and 200 mg) with A and M. Progression from one level to the next was based on prespecified minimum disease stabilization (at least 4/10) and upper limits of specific grade 3-5 treatment-related adverse events (TRAE). RESULTS: The study was able to progress through all 4 dosing levels of sorafenib by the accrual of 40 patients. Thirty-eight (95%) patients had either main portal vein thrombosis or/and extra-hepatic disease. The most common grade 3-5 TRAEs were hand-foot-syndrome (grade 2 and grade 3) in 3 (8%) and transaminitis in 2 (5%) patients, respectively. The plasma concentrations of sorafenib peaked at 600 mg dose, and the concentration threshold of 2400 ng/mL was associated with higher odds of achieving time to exposure (TTE) concentrations >75% centile (odds ratio [OR] = 10.0 [1.67-44.93]; P = .01). The median overall survival for patients without early hepatic decompensation (n = 31) was 8.9 months (95% confidence interval [CI]: 3.2-14.5 months). CONCLUSION: The SAM combination in HCC patients with predominantly unfavorable baseline disease characteristics showed a marked reduction in sorafenib-related side effects. Studies using sorafenib 600 mg per day in this combination along with sorafenib drug level monitoring can be evaluated in further trials.(Trial ID: CTRI/2018/07/014865).


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Liver Neoplasms , Metformin , Antineoplastic Agents/adverse effects , Atorvastatin/therapeutic use , Carcinoma, Hepatocellular/pathology , Humans , Liver Neoplasms/pathology , Metformin/pharmacology , Metformin/therapeutic use , Niacinamide , Phenylurea Compounds/therapeutic use , Sorafenib/therapeutic use , Treatment Outcome
8.
Eur J Clin Pharmacol ; 78(8): 1321-1330, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35652931

ABSTRACT

PURPOSE: Imatinib is a substrate of CYP3A4, ABCB1 and ABCG2, and is known to have wide variability in pharmacokinetics (PK). At the same time, a clear relationship between drug levels and response also exists for imatinib in chronic myeloid leukaemia (CML). Therefore, pharmacogenetic-based dosing of imatinib is an attractive proposition. This study aims to characterize the population pharmacokinetics of imatinib in order to identify significant covariates including pharmacogenetic variants. METHODS: Forty-nine patients with CML were enrolled in the study after being on imatinib for at least 4 consecutive weeks. Steady-state pharmacokinetic sampling was performed either in a sparse (4 samples each, n = 44) or intensive manner (9 samples each, n = 5). An additional pharmacogenetic sample was also collected from all patients. Plasma imatinib levels were estimated using a validated HPLC method. Pharmacogenetic variants were identified using the PharmacoScan array platform. Population pharmacokinetic analysis was carried out using NONMEM v7.2. Seven SNPs within CYP3A4, ABCB1 and ABCG2 genes were evaluated for covariate effect on the clearance of imatinib. RESULTS: Imatinib PK was well characterized using a one-compartment model with zero-order absorption. The clearance and volume of distribution were found to be 10.2 L/h and 389 L respectively. Only SNP rs1128503 of the ABCB1 gene had a small but insignificant effect on imatinib clearance, with a 25% reduction in clearance observed in patients carrying the polymorphism. Twenty-three out of forty-nine patients (47%) carried the polymorphic allele, of whom 17 were heterozygous and six were homozygous. CONCLUSION: Our study conclusively proves that genetic polymorphisms in the CYP3A4 and ABC family of transporters do not have any role in the personalized dosing of imatinib in CML.


Subject(s)
Antineoplastic Agents , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Antineoplastic Agents/pharmacokinetics , Cytochrome P-450 CYP3A/genetics , Humans , Imatinib Mesylate/pharmacokinetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Pharmacogenetics , Polymorphism, Single Nucleotide
9.
J Oncol Pharm Pract ; 28(7): 1641-1644, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35119329

ABSTRACT

INTRODUCTION: Oxaliplatin is a platinum containing alkylating agent commonly used in the management of colorectal cancers. The most common dose-limiting toxicity of oxaliplatin is peripheral neuropathy, which can be severe enough to cause treatment discontinuation. We present a case of dysarthria and laryngopharyngeal dysesthesia (LPD) that developed during the first dose of oxaliplatin, which showed dose-dependent reduction in severity in subsequent cycles. CASE REPORT: A 52-year-old female patient with adenocarcinoma of rectum (pT4N2M0) was prescribed oxaliplatin (130 mg/m2) and capecitabine(2000mg/m2). She developed heaviness in the tongue, slurred speech, jaw pain, perioral paresthesia within 30 min after the end of 3 h infusion of oxaliplatin in the first cycle. The symptoms subsided without any sequelae in two days. However, in the subsequent cycles as the dose of the oxaliplatin was reduced, similar symptoms reappeared but were of reduced in severity. No dysesthesia symptoms were observed in the 4th cycle when the oxaliplatin was administered at 85 mg/m2. MANAGEMENT AND OUTCOME: As and when the patient developed symptoms - slurred speech, jaw pain during the first three cycles, she was managed with inj. Hydrocortisone (100 mg i.v.) and one ampoule of pheniramine (45 mg i.v.). DISCUSSION: Occurrence of laryngopharyngeal dysesthesia due to oxaliplatin does not warrant drug withdrawal, dose titration can be helpful, thereby preventing the drug withdrawal for the patient management.


Subject(s)
Antineoplastic Agents , Colorectal Neoplasms , Antineoplastic Agents/adverse effects , Antineoplastic Combined Chemotherapy Protocols , Capecitabine/therapeutic use , Colorectal Neoplasms/drug therapy , Deoxycytidine , Dysarthria , Female , Fluorouracil , Humans , Middle Aged , Organoplatinum Compounds/adverse effects , Oxaliplatin/adverse effects , Pain/drug therapy , Paresthesia/chemically induced , Paresthesia/drug therapy , Speech
10.
Drug Chem Toxicol ; : 1-14, 2022 Jul 28.
Article in English | MEDLINE | ID: mdl-35899689

ABSTRACT

5-hydroxy-1,4-naphthoquinone (5NQ) or juglone is a bioactive molecule found in walnuts and has shown therapeutic effects in various disease models. Limited information is available regarding the toxicity of 5NQ, thereby limiting the clinical development of this drug. In the present study, oral acute (50, 300 and 2000 mg/kg) and sub-acute toxicity (5, 15 and 50 mg/kg) was assessed in mice to evaluate the safety of 5NQ. The acute toxicity study identified 118 mg/kg as the point-of-departure dose (POD) for single oral administration of 5NQ using benchmark dose modeling (BMD). Repeated administration of 5NQ at doses of 15 and 50 mg/kg/day caused reduction in food consumption and body weight of mice along with alterations in liver and renal function. Histopathological assessment revealed significant damage to hepatic and renal tissues at all doses in the acute toxicity study, and at higher doses of 15 and 50 mg/kg in the sub-acute toxicity study. We observed dose dependent mortality in sub-acute toxicity study and the no observed adverse effect level (NOAEL) was established as < 5 mg/kg/day. Modeling the survival response in sub-acute toxicity study identified 1.74 mg/kg/day as the POD for repeated administration of 5NQ. Serum levels of aspartate aminotransferase (AST) were most sensitive to 5NQ administration with a lower limit of BMD interval (BMDL) of 1.1 × 10-3 mg/kg/day. The benchmark doses reported in the study can be further used to determine a reference dose of 5NQ for human risk assessment.

11.
Int J Cancer ; 149(7): 1495-1511, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34146401

ABSTRACT

Lipocalin 2 is a siderophore-binding protein that regulates iron homeostasis. Lipocalin 2 expression is elevated in multiple tumor types; however, the mechanisms that drive tumor progression upon Lipocalin 2 expression remain unclear. When Lipocalin 2 is over-expressed, it leads to resistance to 5-fluorouracil in colon cancer cell lines in vitro and in vivo by inhibiting ferroptosis. Lipocalin 2 inhibits ferroptosis by decreasing intracellular iron levels and stimulating the expression of glutathione peroxidase4 and a component of the cysteine glutamate antiporter, xCT. The increase in xCT levels is dependent on increased levels of ETS1 in Lipocalin 2 over-expressing cells. Inhibiting Lipocalin 2 function with a monoclonal antibody leads to a decrease in chemo-resistance and transformation in vitro, and a decrease in tumor progression and chemo-resistance in xenograft mouse models. Lipocalin 2 and xCT levels exhibit a positive correlation in human tumor samples suggesting that the pathway we have identified in cell lines is operative in human tumor samples. These results indicate that Lipocalin 2 is a potential therapeutic target and that the monoclonal antibody described in our study can serve as the basis for a potential therapeutic in patients who do not respond to chemotherapy.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Lipocalin-2/metabolism , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Cell Movement , Cell Proliferation , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Humans , Lipocalin-2/genetics , Mice , Mice, Nude , Prognosis , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Am J Transplant ; 20(8): 2044-2057, 2020 08.
Article in English | MEDLINE | ID: mdl-32040239

ABSTRACT

There is an unmet medical need for radiation countermeasures that can be deployed for treatment of exposed individuals during ionizing radiation (IR) accidents or terrorism. Wharton's jelly mesenchymal stem cells (WJ-MSCs) from human umbilical cord have been shown to avoid allorecognition and induce a tissue-regenerating microenvironment, which makes them an attractive candidate for mitigating IR injury. We found that WJ-MSCs protected mice from a lethal dose of IR even when transplanted up to 24 hours after irradiation, and a combination of WJ-MSCs and antibiotic (tetracycline) could further expand the window of protection offered by WJ-MSCs. This combinatorial approach mitigated IR-induced damage to the hematopoietic and gastrointestinal system. WJ-MSCs increased the serum concentration of the cytoprotective cytokines granulocyte colony-stimulating factor (G-CSF) and IL-6 in mice. Knockdown of G-CSF and IL-6 in WJ-MSCs before injection to lethally irradiated mice or transplantation of WJ-MSCs to lethally irradiated Nrf-2 knockout mice significantly nullified the therapeutic protective efficacy. Hence, WJ-MSCs could be a potential cell-based therapy for individuals accidentally exposed to radiation.


Subject(s)
Acute Radiation Syndrome , Mesenchymal Stem Cells , Wharton Jelly , Acute Radiation Syndrome/therapy , Animals , Cell Differentiation , Cells, Cultured , Humans , Mice , Regeneration , Umbilical Cord
13.
J Neurooncol ; 146(3): 477-488, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32020478

ABSTRACT

PURPOSE: Heterogeneity within GBMs and variability of visualized fluorescence combine to confer practical limitations to the technique of optical imaging. A biometric analysis was planned to objectively ascertain and analyse this phenomenon METHODS: 25 adult glioblastoma subjects undergoing resection were prospectively accrued. Biopsies were taken from various parts of the tumor and safe peritumoral zones. White light (WL) and visualized fluorescence was subjectively recorded. Corresponding histopathology [coalescent (C) or infiltrating (I) tumor] and protoporphyrin-IX (PPIX) levels were assayed. RESULTS: WL was very sensitive for detecting tumor. SF was more specific and had high positive predictive value for detecting tumor. WF on the other hand had a poor discriminatory efficacy. Mean PPIX levels were 3.0, 2.01 and 0.16 for SF, WF, and NF respectively. WF had a wide variable range of PPIX levels. Within the coalescent tumor areas, there was a variable distribution of fluorescence (both subjective as well as objective PPIX levels) with only 54% samples showing SF and high PPIX. In seven cases this discordance was noted within the same tumor (biological heterogeneity). CONCLUSIONS: Fluorescence may miss important tumor areas even if objective assessment is used. Histologically similar tumor areas may exhibit contrasting fluorescence properties, a phenomenon which needs further investigation and elucidation of underlying mechanisms which could potentially be manipulated to optimize the utility of fluorescence guidance.


Subject(s)
Brain Neoplasms/diagnostic imaging , Brain Neoplasms/surgery , Glioblastoma/diagnostic imaging , Glioblastoma/surgery , Optical Imaging/methods , Protoporphyrins/analysis , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Prospective Studies , Sensitivity and Specificity
14.
Br J Clin Pharmacol ; 86(6): 1081-1088, 2020 06.
Article in English | MEDLINE | ID: mdl-31925802

ABSTRACT

AIMS: L-asparaginase is an essential medicine in the treatment of pediatric acute lymphoblastic leukemia (ALL) and the quality of generic formulations is an area of concern. We compared nine generic formulations of L-asparaginase available in India with the innovator. METHODS: The quality of formulations was assessed by measuring 72-hour trough asparaginase activity in children with ALL during induction following administration of 10,000 IU/m2 of L-asparaginase. In-vitro analysis of the label claim was assessed by measuring activity of three generic formulations. Liquid chromatography-mass spectrometry (LC/MS) was used to determine the amount of host contaminant proteins (HCPs) in the formulations. RESULTS: Between March 2015 to June 2018, 240 samples from 195 patients were analyzed. The number of samples analyzed ranged from 7-66 per generic brand (median: 18) and seven of the innovator. The proportion of generic formulations that failed to achieve a predefined clinical threshold activity of 50 IU/L ranged from 16.7% (2/12) to 84.9% (28/33) in the highest activity to lowest activity generic respectively. On other hand, all innovator samples had activity greater than 50 IU/L. In-vitro asparaginase activity in the three generic formulations tested ranged from 71.4-74.6% of the label claim (10,000 IU) compared to 93.5% for the innovator. LC/MS analysis of generic 5 identified 25 HCPs with a relative peptide count of 27.1% of the total peptides. CONCLUSIONS: Generic formulations had lower asparaginase activity which raises serious clinical concerns regarding their quality. Until stringent regulatory enforcement improves the quality of these generics, dose adaptive strategies coupled with therapeutic drug monitoring need to be considered.


Subject(s)
Antineoplastic Agents , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Antineoplastic Agents/therapeutic use , Asparaginase/therapeutic use , Child , Escherichia coli , Humans , India , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Retrospective Studies
15.
Bioorg Med Chem Lett ; 30(23): 127561, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32961322

ABSTRACT

Thiazolidinedione (TZD) has been an interesting scaffold due to its proven antidiabetic activity and encouraging findings in anticancer drug discovery. We synthesised benzylidene thiazolidinedione derivatives which exhibited excellent antiproliferative effects in chronic myeloid leukemic cells K562 and the most active compounds 3t and 3x had GI50 value of 0.9 and 0.23 µM respectively. Both the compound was found to arrest the growth of K562 cells in G0/G1 phase in a time and dose dependent manner. Further, western blot analysis revealed that 3t and 3x could also inhibit the expression of cell proliferation markers, PCNA and Cyclin D1 and compound 3x up-regulated apoptosis markers, cleaved PARP1 and activated caspase 3, which could be a possible mechanism for the excellent antiproliferative effects exhibited by these compounds. In vitro combination studies of 3t and 3x with Imatinib found to potentiate the antitumor effects of Imatinib. Further in vivo efficacy in K562 xenografts, of 3t and 3x alone and in combination with Imatinib was found to be promising and far better than control group and combination treatment was found to be more effective as compared to only Imatinib treated or test compound treated animals. Thus, our findings suggest that these compounds are promising antitumor agents and could help to enhance the anticancer effects of Imatinib and other tyrosine kinase inhibitors, when used in combination.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzylidene Compounds/therapeutic use , Imatinib Mesylate/therapeutic use , Neoplasms/drug therapy , Thiazolidinediones/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/toxicity , Antineoplastic Combined Chemotherapy Protocols , Apoptosis/drug effects , Benzylidene Compounds/chemical synthesis , Benzylidene Compounds/toxicity , Cell Line, Tumor , Cell Proliferation/drug effects , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Male , Mice, Nude , Molecular Structure , Structure-Activity Relationship , Thiazolidinediones/chemical synthesis , Thiazolidinediones/toxicity , Xenograft Model Antitumor Assays
16.
Eur J Clin Pharmacol ; 76(10): 1427-1436, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32529316

ABSTRACT

PURPOSE: Patients of non-small cell lung cancer (NSCLC) with brain metastases have limited treatment options. High-dose erlotinib (HDE) and gefitinib (HDG) have been tried in the past. This study investigates the cerebrospinal fluid (CSF) disposition and safety of both, high-dose erlotinib and gefitinib regimens. METHODS: Eleven and nine patients were treated with erlotinib and gefitinib, respectively. All patients received 1 week of standard dose of erlotinib (150 mg OD) or gefitinib (250 mg OD), followed by the high dose (1500 mg weekly for erlotinib and 1250 mg OD for gefitinib) from day 8. Blood and CSF samples were collected on days 7 and 15, 4 h after the morning dose and drug levels determined using LC-MS/MS. Adverse events were documented as per CTCAE 4.03 till day 15. RESULTS: Pulsatile HDE and daily HDG resulted in 1.4- and 1.9-fold increase in CSF levels, respectively. A constant 2% CSF penetration rate was observed across both doses of erlotinib, while for gefitinib the penetration rate for high dose was half that of the standard dose suggesting a nonlinear disposition. Three patients on HDE treatment discontinued treatment after the first dose due to intolerable toxicities, whereas HDG was better tolerated with no treatment discontinuations. Since CSF disposition of gefitinib followed saturable kinetics, a lower dose of 750 mg was found to achieve CSF concentrations comparable to that of the 1250 mg dose. CONCLUSIONS: HDG was better tolerated than HDE. CSF disposition of gefitinib was found to be saturable at a higher dose. Based on these findings, the dose of 750 mg OD should be considered for further evaluation in this setting.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Erlotinib Hydrochloride/administration & dosage , Gefitinib/administration & dosage , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Brain Neoplasms/secondary , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Chromatography, Liquid , Dose-Response Relationship, Drug , Erlotinib Hydrochloride/adverse effects , Erlotinib Hydrochloride/pharmacokinetics , Female , Gefitinib/adverse effects , Gefitinib/pharmacokinetics , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Male , Middle Aged , Tandem Mass Spectrometry
17.
Natl Med J India ; 31(6): 334-338, 2018.
Article in English | MEDLINE | ID: mdl-31397364

ABSTRACT

Background: An 'informed consent' is a legal and ethical requirement for research involving human subjects. Studies assessing the validity of informed consent and determinants of its quality have highlighted problems in consent delivery and comprehension by trial participants. We report the findings of a questionnaire-based survey conducted to understand the quality of informed consent (QuIC) in cancer clinical trials. Methods: The survey was conducted in a single tertiary care cancer centre in India. Patients enrolled in phase 1, 2 or 3 interventional studies were administered the QuIC questionnaire by a trained study coordinator. The QuIC, expressed as knowledge score, was calculated from the proportion of correct responses expressed as a percentage. Results: The mean (SD) knowledge score was 60.46% (1 5.21%). It was considerably higher in industry-sponsored trials compared to investigator-initiated trials (65.32% v. 52.21%, respectively; p<0.001). Faith in the treating oncologist positively influenced the patients' decision to participate in a trial. Nearly 97% of the respondents anticipated better care, while 85% felt that the new drug/procedure would be better than the existing treatment. Free treatment emerged as a strong inducement for patients to take part in clinical trials. Patients were aware of their autonomy, and responses showed that none of the patients were coerced or unduly influenced to participate in clinical trials. Conclusion: Our study revealed important deficiencies in research participants' understanding of essential elements of informed consent. Thorough patient counselling is crucial to minimize 'therapeutic misconception' to preserve the validity of informed consent in cancer trials.


Subject(s)
Clinical Trials as Topic/standards , Health Literacy/statistics & numerical data , Informed Consent , Neoplasms/therapy , Physician-Patient Relations , Adult , Aged , Consent Forms/standards , Cross-Sectional Studies , Female , Humans , India , Male , Middle Aged , Patient Education as Topic , Surveys and Questionnaires/statistics & numerical data , Young Adult
18.
J Labelled Comp Radiopharm ; 60(1): 12-19, 2017 01.
Article in English | MEDLINE | ID: mdl-27813128

ABSTRACT

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


Subject(s)
Antineoplastic Agents, Immunological/chemical synthesis , Iodine Radioisotopes/chemistry , Radiopharmaceuticals/chemical synthesis , Trastuzumab/chemistry , Animals , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/pharmacokinetics , Cell Line, Tumor , Female , Humans , Mammary Neoplasms, Experimental/drug therapy , Mice , Mice, Inbred NOD , Mice, SCID , Protein Binding , Radiopharmaceuticals/adverse effects , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Receptor, ErbB-2/metabolism , Tissue Distribution
19.
AAPS PharmSciTech ; 18(3): 710-720, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27222025

ABSTRACT

Surface modification of liposomes with targeting ligands is known to improve the efficacy with reduced untoward effects in treating infective diseases like visceral leishmaniasis (VL). In the present study, modified ligand (ML), designed by modifying polysaccharide with a long chain lipid was incorporated in liposomes with the objective to target amphotericin B (Amp B) to reticuloendothelial system and macrophages. Conventional liposomes (CL) and surface modified liposomes (SML) were characterized for size, shape, and entrapment efficiency (E.E.). Amp B SML with 3% w/w of ML retained the vesicular nature with particle size of ∼205 nm, E.E. of ∼95% and good stability. SML showed increased cellular uptake in RAW 264.7 cells which could be attributed to receptor-mediated endocytosis. Compared to Amp B solution, Amp B liposomes exhibited tenfold increased safety in vitro in RAW 264.7 and J774A.1 cell lines. Pharmacokinetics and biodistribution studies revealed high t 1/2, area under the curve (AUC)0-24, reduced clearance and prolonged retention in liver and spleen with Amp B SML compared to other formulations. In promastigote and amastigote models, Amp B SML showed enhanced performance with low 50% inhibitory concentration (IC50) compared to Amp B solution and Amp B CL. Thus, due to the targeting ability of ML, SML has the potential to achieve enhanced efficacy in treating VL.


Subject(s)
Amphotericin B/chemistry , Amphotericin B/pharmacology , Leishmaniasis, Visceral/drug therapy , Liposomes/chemistry , Amphotericin B/pharmacokinetics , Animals , Cell Line , Chemistry, Pharmaceutical/methods , Liver/parasitology , Macrophages/parasitology , Mice , Particle Size , Spleen/parasitology , Tissue Distribution
20.
Indian J Med Res ; 143(2): 145-59, 2016 Feb.
Article in English | MEDLINE | ID: mdl-27121513

ABSTRACT

Cellular resistance in tumour cells to different therapeutic approaches has been a limiting factor in the curative treatment of cancer. Resistance to therapeutic radiation is a common phenomenon which significantly reduces treatment options and impacts survival. One of the mechanisms of acquiring resistance to ionizing radiation is the overexpression or activation of various oncogenes like the EGFR (epidermal growth factor receptor), RAS (rat sarcoma) oncogene or loss of PTEN (phosphatase and tensin homologue) which in turn activates the phosphatidyl inositol 3-kinase/protein kinase B (PI3-K)/AKT pathway responsible for radiation resistance in various tumours. Blocking the pathway enhances the radiation response both in vitro and in vivo. Due to the differential activation of this pathway (constitutively activated in tumour cells and not in the normal host cells), it is an excellent candidate target for molecular targeted therapy to enhance radiation sensitivity. In this regard, HIV protease inhibitors (HPIs) known to interfere with PI3-K/AKT signaling in tumour cells, have been shown to sensitize various tumour cells to radiation both in vitro and in vivo. As a result, HPIs are now being investigated as possible radiosensitizers along with various chemotherapeutic drugs. This review describes the mechanisms by which PI3-K/AKT pathway causes radioresistance and the role of HIV protease inhibitors especially nelfinavir as a potential candidate drug to target the AKT pathway for overcoming radioresistance and its use in various clinical trials for different malignancies.


Subject(s)
HIV Protease Inhibitors/therapeutic use , Neoplasms/drug therapy , Neoplasms/radiotherapy , Oncogene Protein v-akt/genetics , Radiation-Sensitizing Agents/therapeutic use , ErbB Receptors/biosynthesis , Humans , Neoplasms/pathology , Oncogene Protein p21(ras)/biosynthesis , Oncogene Protein v-akt/antagonists & inhibitors , PTEN Phosphohydrolase/biosynthesis , Phosphatidylinositol 3-Kinases/genetics , Radiation, Ionizing , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL