Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.037
Filter
1.
Cell Death Dis ; 15(5): 322, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38719798

ABSTRACT

Metastatic dissemination from the primary tumor is a complex process that requires crosstalk between tumor cells and the surrounding milieu and involves the interplay between numerous cellular-signaling programs. Epithelial-mesenchymal transition (EMT) remains at the forefront of orchestrating a shift in numerous cellular programs, such as stemness, drug resistance, and apoptosis that allow for successful metastasis. Till date, there is limited success in therapeutically targeting EMT. Utilizing a high throughput screen of FDA-approved compounds, we uncovered a novel role of the topoisomerase inhibitor, Teniposide, in reversing EMT. Here, we demonstrate Teniposide as a potent modulator of the EMT program, specifically through an IRF7-NMI mediated response. Furthermore, Teniposide significantly reduces the expression of the key EMT transcriptional regulator, Zinc Finger E-Box Binding Homeobox 2 (ZEB2). ZEB2 downregulation by Teniposide inhibited RNA polymerase I (Pol I) activity and rRNA biogenesis. Importantly, Teniposide treatment markedly reduced pulmonary colonization of breast cancer cells. We have uncovered a novel role of Teniposide, which when used at a very low concentration, mitigates mesenchymal-like invasive phenotype. Overall, its ability to target EMT and rRNA biogenesis makes Teniposide a viable candidate to be repurposed as a therapeutic option to restrict breast cancer metastases.


Subject(s)
Breast Neoplasms , Down-Regulation , Epithelial-Mesenchymal Transition , RNA Polymerase I , Teniposide , Zinc Finger E-box Binding Homeobox 2 , Epithelial-Mesenchymal Transition/drug effects , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Female , Zinc Finger E-box Binding Homeobox 2/metabolism , Zinc Finger E-box Binding Homeobox 2/genetics , Cell Line, Tumor , Down-Regulation/drug effects , RNA Polymerase I/metabolism , Teniposide/pharmacology , Animals , Mice , Gene Expression Regulation, Neoplastic/drug effects
2.
Neuro Oncol ; 26(4): 724-734, 2024 04 05.
Article in English | MEDLINE | ID: mdl-38037691

ABSTRACT

BACKGROUND: Studies on the efficacy of rituximab in primary CNS lymphoma (PCNSL) reported conflicting results. Our international randomized phase 3 study showed that the addition of rituximab to high-dose methotrexate, BCNU, teniposide, and prednisolone (MBVP) in PCNSL was not efficacious in the short term. Here we present long-term results after a median follow-up of 82.3 months. METHODS: One hundred and ninety-nine eligible newly diagnosed, nonimmunocompromised patients with PCNSL aged 18-70 years with WHO performance status 0-3 was randomized between treatment with MBVP chemotherapy with or without rituximab, followed by high-dose cytarabine consolidation in responding patients, and reduced-dose WBRT in patients aged ≤ 60 years. Event-free survival was the primary endpoint. Overall survival rate, neurocognitive functioning (NCF), and health-related quality of life (HRQoL) were additionally assessed, with the IPCG test battery, EORTC QLQ-C30 and QLQ-BN20 questionnaires, respectively. RESULTS: For event-free survival, the hazard ratio was 0.85, 95% CI 0.61-1.18, P = .33. Overall survival rate at 5 years for MBVP and R-MBVP was 49% (39-59) and 53% (43-63) respectively. In total, 64 patients died in the MBVP arm and 55 in the R-MBVP arm, of which 69% were due to PCNSL. At the group level, all domains of NCF and HRQoL improved to a clinically relevant extent after treatment initiation, and remained stable thereafter up to 60 months of follow-up, except for motor speed which deteriorated between 24 and 60 months. Although fatigue improved initially, high levels persisted in the long term. CONCLUSIONS: Long-term follow-up confirms the lack of added value of rituximab in addition to MBVP and HD-cytarabine for PCNSL.


Subject(s)
Central Nervous System Neoplasms , Lymphoma , Humans , Methotrexate/therapeutic use , Rituximab/therapeutic use , Teniposide/therapeutic use , Carmustine/therapeutic use , Lymphoma/therapy , Prednisolone/therapeutic use , Quality of Life , Combined Modality Therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Central Nervous System Neoplasms/pathology , Cytarabine/therapeutic use
3.
BMC Cancer ; 23(1): 746, 2023 Aug 11.
Article in English | MEDLINE | ID: mdl-37568079

ABSTRACT

BACKGROUND: This study aimed to compare the efficacy and safety of high-dose methotrexate (HD-MTX) versus teniposide (TEN) in patients with newly diagnosed immunocompetent primary central nervous system lymphomas (PCNSLs). METHODS: The study included immunocompetent, adult patients with newly diagnosed PCNSL at 22 centers in China from 2007 to 2016. The patients received HD-MTX or TEN as first-line induction therapy. The objective response rate, progression-free survival, and overall survival were analyzed for each patient cohort. RESULTS: A total of 96 patients were eligible: 62 received HD-MTX, while 34 received teniposide. The overall response rate was 73.2% and 72.7% in the MTX and the TEN cohorts, respectively (P = 0.627). The median progression-free survival was 28.4 months [95% confidence interval (CI): 13.7-51.2] in the MTX cohort and 24.3 months (95% CI: 16.6-32.1) in the TEN cohort (P = 0.75). The median overall survival was 31 months (95% CI: 26.8-35.2) in the MTX cohort and 32 months (95% CI: 27.6-36.4) in the TEN cohort (P = 0.77). The incidence of any grade of coagulopathy/deep-vein thrombosis and gastrointestinal disorders was significantly higher in the MTX cohort than in the TEN cohort; no significant difference was found in the incidence of other adverse events between the two cohorts. CONCLUSIONS: This was the first multicenter study using TEN as the main agent compared with HD-MTX in newly diagnosed primary CNS lymphoma. The TEN-based regimen was non-inferior to the HD-MTX-based regimen with similar overall responses. CLASSIFICATION OF EVIDENCE: This study provided Class III evidence that the teniposide-based regimen was non-inferior to high-dose methotrexate - based regimen with similar overall responses and long-time survival in immunocompetent patients with PCNSL.


Subject(s)
Central Nervous System Neoplasms , Lymphoma , Adult , Humans , Methotrexate/therapeutic use , Teniposide/therapeutic use , Induction Chemotherapy , Retrospective Studies , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Central Nervous System Neoplasms/pathology , Central Nervous System
4.
Comput Biol Med ; 163: 107078, 2023 09.
Article in English | MEDLINE | ID: mdl-37356294

ABSTRACT

BACKGROUND: TP53 mutation and hypoxia play an essential role in cancer progression. However, the metabolic reprogramming and tumor microenvironment (TME) heterogeneity mediated by them are still not fully understood. METHODS: The multi-omics data of 32 cancer types and immunotherapy cohorts were acquired to comprehensively characterize the metabolic reprogramming pattern and the TME across cancer types and explore immunotherapy candidates. An assessment model for metabolic reprogramming was established by integration of multiple machine learning methods, including lasso regression, neural network, elastic network, and survival support vector machine (SVM). Pharmacogenomics analysis and in vitro assay were conducted to identify potential therapeutic drugs. RESULTS: First, we identified metabolic subtype A (hypoxia-TP53 mutation subtype) and metabolic subtype B (non-hypoxia-TP53 wildtype subtype) in hepatocellular carcinoma (HCC) and showed that metabolic subtype A had an "immune inflamed" microenvironment. Next, we established an assessment model for metabolic reprogramming, which was more effective compared to the traditional prognostic indicators. Then, we identified a potential targeting drug, teniposide. Finally, we performed the pan-cancer analysis to illustrate the role of metabolic reprogramming in cancer and found that the metabolic alteration (MA) score was positively correlated with tumor mutational burden (TMB), neoantigen load, and homologous recombination deficiency (HRD) across cancer types. Meanwhile, we demonstrated that metabolic reprogramming mediated a potential immunotherapy-sensitive microenvironment in bladder cancer and validated it in an immunotherapy cohort. CONCLUSION: Metabolic alteration mediated by hypoxia and TP53 mutation is associated with TME modulation and tumor progression across cancer types. In this study, we analyzed the role of metabolic alteration in cancer and propose a predictive model for cancer prognosis and immunotherapy responsiveness. We also explored a potential therapeutic drug, teniposide.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Teniposide , Tumor Microenvironment , Hypoxia/genetics , Mutation , Tumor Suppressor Protein p53/genetics
7.
Chembiochem ; 23(20): e202200284, 2022 10 19.
Article in English | MEDLINE | ID: mdl-36040838

ABSTRACT

Target engagement and the biodistribution of exogenously administered small molecules is rarely homogenous. Methods to determine the biodistribution at the cellular level are limited by the ability to detect the small molecule and simultaneously identify the cell types or tissue structures with which it is associated. The highly multiplexed nature of mass cytometry could facilitate these studies provided a heavy isotope label was available in the molecule of interest. Here we show it is possible to append a tellurophene to a known chemotherapeutic, teniposide, to follow this molecule in vivo. A semi-synthetic approach offers an efficient route to the teniposide analogue which is found to have similar characteristics when compared with the parent teniposide in vitro. Using mass cytometry we find the teniposide analogue has significant nonspecific binding to cells. In vivo the tellurium bearing teniposide produces the expected DNA damage in a PANC-1 xenograft model. The distribution of Te in the tissue is near the limits of detection and further work will be required to characterize the localization of this analogue with respect to cell type distributions.


Subject(s)
Tellurium , Teniposide , Humans , Tissue Distribution , DNA Damage
8.
J Immunother Cancer ; 10(8)2022 08.
Article in English | MEDLINE | ID: mdl-36002188

ABSTRACT

BACKGROUND: Emerging evidence indicates that the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) axis plays a pivotal role in intrinsic antitumor immunity. Previous studies demonstrate that the conventional chemotherapy agent, teniposide, effectively promotes the therapeutic efficacy of programmed cell death protein-1 antibody (PD-1 Ab) through robust cGAS-STING activation. Unfortunately, the cGAS expression of tumor cells is reported to be severely suppressed by the hypoxic status in solid tumor. Clinically, enhancing chemotherapy-induced, DNA-activated tumor STING signaling by alleviating tumor hypoxia might be one possible direction for improving the currently poor response rates of patients with hepatocellular carcinoma (HCC) to PD-1 Ab. METHODS: Teniposide was first screened out from several chemotherapy drugs according to their potency in inducing cGAS-STING signaling in human HCC cells. Teniposide-treated HCC cells were then cultured under hypoxia, normoxia or reoxygenation condition to detect change in cGAS-STING signaling. Next, oxaliplatin/teniposide chemotherapy alone or combined with hyperbaric oxygen (HBO) therapy was administered on liver orthotopic mouse tumor models, after which the tumor microenvironment (TME) was surveyed. Lastly, teniposide alone or combined with HBO was performed on multiple mouse tumor models and the subsequent anti-PD-1 therapeutic responses were observed. RESULTS: Compared with the first-line oxaliplatin chemotherapy, teniposide chemotherapy induced stronger cGAS-STING signaling in human HCC cells. Teniposide-induced cGAS-STING activation was significantly inhibited by hypoxia inducible factor 1α in an oxygen-deficient environment in vitro and the inhibition was rapidly removed via effective reoxygenation. HBO remarkably enhanced the cGAS-STING-dependent tumor type Ⅰ interferon and nuclear factor kappa-B signaling induced by teniposide in vivo, both of which contributed to the activation of dendritic cells and subsequent cytotoxic T cells. Combined HBO with teniposide chemotherapy improved the therapeutic effect of PD-1 Ab in multiple tumor models. CONCLUSIONS: By combination of two therapies approved by the Food and Drug Administration, we safely stimulated an immunogenic, T cell-inflamed HCC TME, leading to further sensitization of tumors to anti-PD-1 immunotherapy. These findings might enrich therapeutic strategies for advanced HCC andwe can attempt to improve the response rates of patients with HCC to PD-1 Ab by enhancing DNA-activated STING signaling through effective tumor reoxygenation.


Subject(s)
Carcinoma, Hepatocellular , Hyperbaric Oxygenation , Liver Neoplasms , Animals , Antibodies , Carcinoma, Hepatocellular/drug therapy , Humans , Hypoxia , Liver Neoplasms/drug therapy , Membrane Proteins , Mice , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Oxaliplatin , Oxygen , Teniposide , Tumor Microenvironment , United States
9.
Comput Intell Neurosci ; 2022: 1098394, 2022.
Article in English | MEDLINE | ID: mdl-35515501

ABSTRACT

Background: Gastric cancer is the second most frequent cause of cancer death worldwide, although much geographical variation in incidence exists. Prevention and personalized treatment are regarded as the best options to reduce gastric cancer mortality rates (Hartgrink et al., 2009). Numerous studies have suggested that Notch1 and its ligands are overexpressed in gastric cancer, and its knockdown can inhibit the proliferation and survival of gastric cancer cells. Objective: To investigate the effect of Notch1 on the stemness and drug sensitivity of human gastric cancer SGC-7901 cells. Methods: Highly expressed Notch1 intracellular domain (NICD1) and Notch1-shRNA lentiviral expression vector were used to infect human gastric cancer SGC-7901 cells cultured in vitro, and western blot and immunofluorescence staining were used to identify highly expressed NICD and Notch1 silenced cells. The percentage of CD133+ cells was analyzed by flow cytometry, the expression of nestin and CFAP by immunofluorescence staining, the formation rate of tumor cell spheres and the tumorigenicity of SCID mice in vivo, and the regulation of cell stemness by Notch1. The sensitivity of each group of cells to the chemotherapeutic drugs teniposide (VM-26) and carmustine (BCNU) was also detected by the MTT method. Results: The stemness phenotype of tumor cells with the increased NICD expression was enhanced, such as an increased proportion of CD133+ cells, enhanced nestin expression, decreased GFAP expression, increased tumor cell sphere formation rate and tumorigenic rate of SCID mice implantation, and decreased sensitivity to VM-26 and BCNU. In contrast, the stemness phenotype of tumor cells with downregulated Notch1 gene expression was significantly suppressed, while the sensitivity to VM-26 and BCNU was increased. Conclusion: High Notch1 expression increased the stemness of SGC-7901 cells and decreased the sensitivity of SGC-7901 cells to chemotherapeutic drugs.


Subject(s)
Stomach Neoplasms , Animals , Carmustine/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, SCID , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Nestin/genetics , Nestin/metabolism , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Signal Transduction , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Teniposide/metabolism
10.
Hematology Am Soc Hematol Educ Program ; 2021(1): 682-688, 2021 12 10.
Article in English | MEDLINE | ID: mdl-34889374

ABSTRACT

Systemic light chain (AL) amyloidosis is a protein misfolding disorder characterized by the deposition of abnormal immunoglobulin light chains in fibrillary aggregates, resulting in end-organ damage. Several unique challenges face treating physicians, including delayed diagnosis, advanced vital organ involvement, and morbidity with treatment. Aggressive supportive care and risk-adapted application of plasma cell-directed therapies are the cornerstones of management. The therapeutic revolution in multiple myeloma will likely further expand the arsenal against plasma cells. Careful investigation of these agents will be critical to establish their role in this fragile population. The promise of fibril-directed therapies to restore organ function remains despite early disappointments. In this review, we discuss new therapies to tackle AL amyloidosis using a case-based approach.


Subject(s)
Immunoglobulin Light-chain Amyloidosis/therapy , Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cyclophosphamide/therapeutic use , Dexamethasone/therapeutic use , Disease Management , Humans , Immunoglobulin Light Chains/analysis , Immunoglobulin Light-chain Amyloidosis/diagnosis , Immunoglobulin Light-chain Amyloidosis/pathology , Male , Middle Aged , Teniposide/therapeutic use
11.
Ann Palliat Med ; 10(11): 11798-11807, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34872304

ABSTRACT

BACKGROUND: Teniposide, as a more potent inhibitor of topoisomerase II compared with etoposide, shows less damage on hematopoietic stem cells. Few data are available on teniposide in hematopoietic stem cell transplantation (HSCT) for high-risk or refractory recurrent hematopoietic malignant diseases, particularly for acute myeloid leukemia (AML). METHODS: A retrospective single arm study was conducted to confirm the feasibility of teniposide (300 mg/m2) -intensified HSCT in the treatment of high-risk or refractory recurrent hematopoietic malignant disease by analysing the outcomes of 32 patients, who received transplantation between January 2016 and December 2018. Univariate and multivariate analyses were performed to evaluate prognostic factors of the endpoints. Statistically significant factors (P<0.05) in multivariate analyses were regarded to be predictive. RESULTS: All patients achieved myeloid engraftment at a median of 13 days (range, 9-28 days), platelet engraftment at 15.5 days (range, 6-142 days), with a cumulative incidence (CI) of platelet engraftment of 93.75%±0.26%. The CI of grade II-IV acute graft versus host disease (aGVHD) was 43.75%±0.80% and that of grade III-IV aGVHD 12.50%±0.35%. The CI of chronic (c)GVHD was 74.07%±0.82% and that of extensive cGVHD 33.33%±0.87%. The CI of relapse was 35.03%±0.76%. The one-year probability of overall survival (OS) was 62.50%±0.09%, while 2-year OS was 46.90%±0.09%, and those of 1- and 2-year leukemia-free-survival (LFS) were 56.30%±0.09% and 46.90%±0.09%, respectively. Generally, the OS and LFS until the end of our follow up were 43.50%±0.09% and 34.80%±0.11%, respectively. The probability of GVHD-free and relapse-free survival (GRFS) was 24.60%±0.08%. Multivariate analysis indicated that the probability of OS was significantly lower in patients with a disease duration of more than 280 days before receiving HSCT and in those with fewer mononuclear cells. For LFS, other than the above two factors, failure to achieve complete response (CR) before HSCT was another independent risk factor. Similarly, the probability of GRFS was significantly lower in patients with longer disease duration (≥280 days) and those receiving stem cells from female donors. CONCLUSIONS: For patients with high-risk or refractory recurrent hematopoietic malignant disease, teniposide-based conditioning regimens followed by allo-HSCT can be considered as an alternative therapy with encouraging prognoses.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Antilymphocyte Serum/therapeutic use , Female , Graft vs Host Disease/prevention & control , Humans , Recurrence , Retrospective Studies , Teniposide
12.
Int J Biol Sci ; 17(13): 3554-3572, 2021.
Article in English | MEDLINE | ID: mdl-34512165

ABSTRACT

Background: Alcohol consumption increases the risk of hepatocellular carcinoma (HCC), and associated with a high mortality rate and poor prognosis. N6-methyladenosine (m6A) methylations play key roles in tumorigenesis and progression. However, our current knowledge about m6A in alcohol-related HCC (A-HCC) remains elucidated. Herein, the authors construct an integrative m6A model based on A-HCC subtyping and mechanism exploration workflow. Methods: Based on the m6A expressions of A-HCC and in vivo experiment, different prognosis risk A-HCC subtypes are identified. Meanwhile, multiple interdependent indicators of prognosis including patient survival rate, clinical pathological prognosis and immunotherapy sensitivity. Results: The m6A model includes LRPPRC, YTHDF2, KIAA14219, and RBM15B, classified A-HCC patients into high/low-risk subtypes. The high-risk subtype compared to the low-risk subtype showed phenotypic malignancy, poor prognosis, immunosuppression, and activation of tumorigenesis and proliferation-related pathways, including the E2F target, DNA repair, and mTORC1 signalling pathways. The expression of Immunosuppressive cytokines DNMT1/EZH2 was up-regulated in A-HCC patients, and teniposide may be a potential therapeutic drug for A-HCC. Conclusion: Our model redefined A-HCC prognosis risk, identified potential m6As linking tumour progress and immune regulations and selected possible therapy target, thus promoting understanding and clinical applications about A-HCC.


Subject(s)
Adenosine/analogs & derivatives , Carcinoma, Hepatocellular/metabolism , Gene Expression Regulation, Neoplastic , Liver Neoplasms/metabolism , Nomograms , Adenosine/metabolism , Alcohol Drinking/adverse effects , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/etiology , Case-Control Studies , DNA Methylation , Drug Screening Assays, Antitumor , Humans , Liver Neoplasms/diagnosis , Liver Neoplasms/drug therapy , Liver Neoplasms/etiology , Mice, Inbred C57BL , Teniposide/therapeutic use
13.
Eur J Haematol ; 106(4): 563-573, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33496996

ABSTRACT

OBJECTIVE: There are limited data on the efficacy and tolerability of VCD chemotherapy in transplant-non-eligible (TNE) newly diagnosed myeloma (NDMM) patients. In this retrospective study, we set out to evaluate this triplet combination in this setting across Thames Valley Cancer Network (UK). METHODS: The primary end point was overall response rate (ORR). Secondary outcomes included event-free survival (EFS), overall survival (OS) and adverse events (AEs). RESULTS: In a total cohort of 158 patients, ORR for total cohort was 72.1%. Median EFS was 10.5 months, and for subgroups by age (<75:11.7 vs ≥75:10.3 months, P = .124), by Charlson Co-morbidity Index (CCI) (<5:11.1 vs ≥5:8.2 months, P = .345). The 4-month landmark analysis showed the following median EFS results: by cumulative bortezomib dose (≥26 mg/m2 : 9.0 months vs <26 mg/m2 : 6.4, P = .13), by cumulative cyclophosphamide dose (≥7000 mg: 9.2 vs <7000 mg: 7.0 months, P = .02) and by cumulative dexamethasone dose (>600 mg: 7.8 vs ≤600 mg: 8.3 months, P = .665). Median OS was 46.9 months. The incidence rate of AE was as follows: any grade (76.8%), ≥G3 (27.1%), ≥G3 haematological AEs (7.9%), any grade infections (31.1%) and ≥G3 infections (11.9%). CONCLUSION: This study demonstrated a good ORR achieved from fixed duration VCD, which was reasonably well tolerated. This was followed by modest median EFS. We envisage that the latter may be improved in this patient group with the use of a higher cumulative bortezomib dose (≥26 mg/m2 ) which showed a trend for improved EFS although without statistical significance (P = .13), and with the use of a higher cumulative cyclophosphamide doses (≥7000 mg, P = .02), subject to tolerability and close monitoring.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Multiple Myeloma/drug therapy , Multiple Myeloma/epidemiology , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Clinical Decision-Making , Cyclophosphamide/adverse effects , Cyclophosphamide/therapeutic use , Dexamethasone/adverse effects , Dexamethasone/therapeutic use , Disease Management , Female , Health Care Surveys , Humans , Male , Middle Aged , Multiple Myeloma/diagnosis , Multiple Myeloma/mortality , Prognosis , Teniposide/adverse effects , Teniposide/therapeutic use , Treatment Outcome , United Kingdom/epidemiology
14.
Leuk Res ; 99: 106476, 2020 12.
Article in English | MEDLINE | ID: mdl-33171301

ABSTRACT

Cytogenetic abnormalities are a recognized factor in the pathogenesis of multiple myeloma (MM). While chromosomal translocations involving the IGH gene have been investigated and reported, the implications of deletions or amplifications in the IGH gene have been less frequently examined. We conducted a retrospective analysis of 260 patients with MM from Northern Israel. Fluorescent in situ hybridization (FISH) analysis of separated CD-138 positive cells was done on bone marrow samples collected between 2016 and 2018. We used IGH break apart probes to identify IGH abnormalities and performed statistical analysis of clinical and prognostic features, comparing the different cytogenetic groups. Deletions in the variable region of the IGH (IGHv) were found in 17.3 % (n = 45) of patients and correlated with significantly worse progression free survival (PFS) after two years of follow up (p = 0.008), as well as with a worse response to 1st line treatment (p = 0.037). The median PFS was 7.1 and 17.7 months in patients with and without IGHv deletion, respectively. PFS differences remained significant (p = 0.017) in subgroup analysis of patients with high-risk cytogenetics (n = 108, 19 with IGHv deletion). Overall survival was not significantly different in the two groups. Constant region (IGHc) amplifications, were less frequently found (6.15 %, n = 16), yet significantly correlated with worse PFS after two years of follow up (p = 0.023). This difference remained valid in the high-risk subgroup (p = 0.001). In Conclusion, we identified that deletion of the IGH variable region and amplification in the IGH constant region, are both associated with poor prognosis and inferior outcome in MM.


Subject(s)
Genes, Immunoglobulin Heavy Chain , Multiple Myeloma/genetics , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cyclophosphamide/administration & dosage , Dexamethasone/administration & dosage , Female , Follow-Up Studies , Gene Amplification , Gene Deletion , Humans , Immunoglobulin Constant Regions/genetics , Immunoglobulin Variable Region/genetics , In Situ Hybridization, Fluorescence , Israel/epidemiology , Male , Middle Aged , Multiple Myeloma/drug therapy , Multiple Myeloma/mortality , Plasma Cells/ultrastructure , Prognosis , Progression-Free Survival , Retrospective Studies , Teniposide/administration & dosage , Treatment Outcome
15.
J Med Chem ; 63(6): 2877-2893, 2020 03 26.
Article in English | MEDLINE | ID: mdl-32084316

ABSTRACT

As an FDA-approved drug, teniposide, was utilized in cancer treatment but was accompanied by a strong side effect in long-term clinical trials. This work discovered potential candidate drugs with low toxicity by modifying the molecule structure of teniposide through a structure-guided drug design approach. The IC50 value of novel 4,6-O-thenylidene-ß-d-glucopyranoside-(2″-acetamido, 3″-acetyl-di-S-5-fluorobenzothizole/5-fluorobenzoxazole)-4'-demethylepipodophyllotoxin (compounds 15 and 16) was 120.4-125.1 µM, which was significantly improved by around 10 times more than teniposide (11.5-22.3 µM) against healthy human cells (i.e., HL-7702, H8, MRC-5, and HMEC). In vivo studies demonstrated compounds 15 and 16 significantly suppressed the tumor growth in the HepG2 cell xenograft model without exhibiting obvious toxicity (LD50 values of 208.45 and 167.52 mg/kg), which was lower than that of teniposide (LD50 = 46.12 mg/kg). Compounds 15 and 16 caused mild γH2AX phosphorylation for low DNA toxicity and less inhibition of PI3K/Akt. Compounds 15 and 16 might be potential antitumor drugs with low toxicity.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Podophyllotoxin/analogs & derivatives , Teniposide/analogs & derivatives , Teniposide/pharmacology , Animals , Antineoplastic Agents/toxicity , Cell Line , DNA Damage/drug effects , Hep G2 Cells , Humans , Male , Mice, Inbred C57BL , Molecular Docking Simulation , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/chemistry , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors/toxicity , Podophyllotoxin/chemistry , Podophyllotoxin/pharmacology , Podophyllotoxin/toxicity , Teniposide/toxicity
16.
J Clin Invest ; 129(11): 4850-4862, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31408442

ABSTRACT

Checkpoint blockade antibodies have been approved as immunotherapy for multiple types of cancer, but the response rate and efficacy are still limited. There are few immunogenic cell death (ICD)-inducing drugs available that can kill cancer cells, enhance tumor immunogenicity, increase the in vivo immune infiltration, and thereby boosting a tumor response to immunotherapy. So far, the ICD markers have been identified as the few immuno-stimulating characteristics of dead cells, but whether the presence of such ICD markers on tumor cells translates into enhanced antitumor immunity in vivo is still investigational. To identify anticancer drugs that could induce tumor cell death and boost T cell response, we performed drug screenings based on both an ICD reporter assay and T cell activation assay. We identified that teniposide, a DNA topoisomerase II inhibitor, could induce high mobility group box 1 (HMGB1) release and type I interferon signaling in tumor cells, and teniposide-treated tumor cells could activate antitumor T cell response both in vitro and in vivo. Mechanistically, teniposide induced tumor cell DNA damage and innate immune signaling including NF-κB activation and STING-dependent type I interferon signaling, both of which contribute to the activation of dendritic cells and subsequent T cells. Furthermore, teniposide potentiated the antitumor efficacy of anti-PD1 on multiple types of mouse tumor models. Our findings showed that teniposide could trigger tumor immunogenicity, and enabled a potential chemo-immunotherapeutic approach to potentiate the therapeutic efficacy of anti-PD1 immunotherapy.


Subject(s)
Immunity, Cellular/drug effects , Membrane Proteins/immunology , Neoplasm Proteins/immunology , Neoplasms, Experimental/drug therapy , Nucleotidyltransferases/immunology , Signal Transduction/drug effects , Teniposide/pharmacology , Topoisomerase II Inhibitors/pharmacology , Animals , Cell Line, Tumor , Female , HEK293 Cells , Humans , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasm Proteins/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Nucleotidyltransferases/genetics , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Xenograft Model Antitumor Assays
17.
J Cancer Res Clin Oncol ; 145(9): 2343-2355, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31280348

ABSTRACT

PURPOSE: Combinations of bortezomib (Velcade), cyclophosphamide and dexamethasone have shown significant efficacy and safety for patients of newly diagnosed multiple myeloma (NDMM). In this study, we compared the efficacy and safety of modified VCD regimens with novel changes in bortezomib dose and schedule for NDMM. METHODS: Eighty-five NDMM patients from multiple centers were randomly assigned to a high-dose (1.6 mg/m2) (group A) or a low-dose (1.3 mg/m2) (group B) bortezomib, administrated on days 1, 6, 11, and 16 subcutaneously in a 4-week cycle for nine cycles, combined with 40 mg dexamethasone on bortezomib days and cyclophosphamide 300 mg/m2 on days 1-3 intravenously. RESULTS: After four cycles, complete response (CR) or better in group A (43.6%) was higher than that in group B (12.8%) (P = 0.002). During induction, for patients with R-ISS stage III, the CR or better rate in group A was superior to that in group B (P = 0.01). Of patients < 65, the CR or better rate of group A was superior to that of group B (P = 0.004). Rapid onset of CR occurred in group A (P < 0.01). Meanwhile, rate of 3-4 diarrhea was higher in group A (P = 0.03), which caused higher rate of dose reduction for patients ≥ 65 (P = 0.041). No significant difference between the two groups in PFS and OS. CONCLUSIONS: The studied high-dose VCD as induction regimen had an improved CR rate, especially in patients < 65 or with R-ISS stage III, and is feasible for young and high-risk patients. Trial registration ClinicalTrials.gov: NCT02086942.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Bortezomib/administration & dosage , Cyclophosphamide/administration & dosage , Dexamethasone/administration & dosage , Multiple Myeloma/drug therapy , Adult , Aged , Aged, 80 and over , Bortezomib/adverse effects , Cyclophosphamide/adverse effects , Dexamethasone/adverse effects , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Injections, Subcutaneous , Male , Middle Aged , Multiple Myeloma/diagnosis , Teniposide/administration & dosage , Treatment Outcome
18.
BMC Res Notes ; 12(1): 446, 2019 Jul 22.
Article in English | MEDLINE | ID: mdl-31331386

ABSTRACT

OBJECTIVE: We explored detrimental effects of VCD on non-ovarian tissues such as kidneys and liver 14 days post-drug administration. Twelve rats were randomly assigned into two groups. In VCD group, rats received 160 mg/kgbw VCD intraperitoneally for 15 consequent days. Control rats were injected with VCD-free normal saline. At the respective time point, rats were euthanized, blood and tissue samples were collected. H&E staining was performed to evaluate pathological changes. Serum level of ALT, AST, creatinine and urea were also measured. RESULTS: Histological analysis revealed hyperemia and follicular atresia in the ovaries, indicating successful POF induction in rats. In renal tissue, extensive tubular necrosis, focal hemorrhage, hyaline casts, and interstitial nephritis were observed. Analysis of hepatic tissue showed numerous hemorrhagic foci, chronic cholangitis, and hepatocyte necrosis, indicating apparent VCD toxicity of both hepatic and renal tissues. The biochemical evaluation revealed a tendency of increase in ALT, AST, creatinine, and Urea in VCD-treated rats; however, the values did not reach significant level. In conclusion, the induction of POF in rats by VCD correlates with renal and hepatic damages. Commensurate with data from this study, any conclusions from experiments based on VCD-induced premature ovarian failure rats should be reported with caution.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Kidney/drug effects , Liver/drug effects , Ovary/drug effects , Alanine Transaminase/blood , Animals , Antineoplastic Combined Chemotherapy Protocols/toxicity , Aspartate Aminotransferases/blood , Creatinine/blood , Cyclophosphamide/administration & dosage , Cyclophosphamide/toxicity , Dexamethasone/administration & dosage , Dexamethasone/toxicity , Female , Injections, Intraperitoneal , Kidney/pathology , Liver/pathology , Ovary/pathology , Primary Ovarian Insufficiency/chemically induced , Primary Ovarian Insufficiency/diagnosis , Random Allocation , Rats, Wistar , Teniposide/administration & dosage , Teniposide/toxicity
19.
Leuk Lymphoma ; 60(12): 2890-2898, 2019 12.
Article in English | MEDLINE | ID: mdl-31282794

ABSTRACT

This phase-II study assessed activity and toxicity of substituting conventional doxorubicin with nonpegylated liposomal doxorubicin in the conventional ABVD regimen for the treatment of elderly or cardiopathic patients with HL. Stage I-IIA and IIB-IV patients were treated with three courses of MBVD plus radiotherapy, or six courses of MBVD, respectively, plus radiotherapy limited to bulky or residual disease areas. The primary endpoints were CR rate and the rate of cardiac events. Forty-seven patients were enrolled. Median age was 75 years, 13 had stage I-II disease. Overall, CR was achieved by 36 patients (77%, 95% CI: 62-88), 100% and 68% in stage I-II and III-IV, respectively. With a median follow-up of 40 months (IQR: 36-45). Three-year overall survival (OS) and progression-free survival (PFS) were 70% and 43%, respectively. Cardiac events grades 3-5 were reported in two patients. In conclusion, MBVD's activity and safety profile was comparable to historical ABVD data.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Heart Diseases/complications , Hodgkin Disease/complications , Hodgkin Disease/drug therapy , Age Factors , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cardiotoxicity/etiology , Carmustine/adverse effects , Carmustine/therapeutic use , Combined Modality Therapy , Female , Heart Diseases/diagnosis , Hodgkin Disease/diagnosis , Hodgkin Disease/mortality , Humans , Male , Methotrexate/adverse effects , Methotrexate/therapeutic use , Methylprednisolone/adverse effects , Methylprednisolone/therapeutic use , Neoplasm Staging , Prognosis , Recurrence , Remission Induction , Survival Analysis , Teniposide/adverse effects , Teniposide/therapeutic use , Treatment Outcome
20.
Eur J Haematol ; 103(3): 247-254, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31231833

ABSTRACT

OBJECTIVES: At our center, patients with multiple myeloma (MM) were treated upfront with bortezomib, cyclophosphamide, and dexamethasone (VCD) until cyclophosphamide was replaced with lenalidomide in the combination (VRD). These treatments have never been compared head-to-head in large real-life patient material. METHOD: A retrospective analysis of patients treated with VRD and VCD in the first line, both with and without subsequent high-dose treatment (HDT) and autologous stem cell transplantation. A total of 681 patients were included, 117 receiving VRD (71 with, 46 without HDT) and 564 receiving VCD (351 with, 213 without HDT). RESULTS: Overall response rate (≥partial response) was higher with VRD compared to VCD in the entire VRD group (98% vs 88%, P < 0.001) and in the non-HDT group (98% vs 79%, P < 0.001). Progression-free survival (PFS) at 18 months was longer with VRD compared to VCD in the entire VRD group, the non-HDT group and the HDT group (88% vs 63%, 82% vs 32% and 91% vs 73%, respectively). Overall survival at 18 months was better for VRD-treated patients in the entire VRD group (95% vs 89%, P = 0.048). CONCLUSION: Upfront VRD gives better responses and longer PFS compared to VCD in MM patients with or without subsequent HDT.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Multiple Myeloma/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bortezomib/administration & dosage , Cyclophosphamide/adverse effects , Cyclophosphamide/therapeutic use , Dexamethasone/administration & dosage , Dexamethasone/adverse effects , Dexamethasone/therapeutic use , Female , Humans , Lenalidomide/administration & dosage , Male , Middle Aged , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Prognosis , Proportional Hazards Models , Survival Analysis , Teniposide/adverse effects , Teniposide/therapeutic use , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...