Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Cancer Invest ; 42(4): 319-332, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38695671

ABSTRACT

Glioblastoma multiforme (GBM), is a frequent class of malignant brain tumors. Epigenetic therapy, especially with synergistic combinations is highly paid attention for aggressive solid tumors like GBM. Here, RSM optimization has been used to increase the efficient arrest of U87 and U251 cell lines due to synergistic effects. Cell lines were treated with SAHA, 5-Azacytidine, GSK-126, and PTC-209 individually and then RSM was used to find most effective combinations. Results showed that optimized combinations significantly reduce cell survival and induce cell cycle arrest and apoptosis in both cell lines. Expression of cyclin B1 and cyclin D1 were decreased while caspase3 increased expression.


Subject(s)
Apoptosis , Drug Synergism , Epigenesis, Genetic , Glioblastoma , Humans , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , Cell Line, Tumor , Apoptosis/drug effects , Epigenesis, Genetic/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Azacitidine/pharmacology , Azacitidine/administration & dosage , Cell Survival/drug effects , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Cycle Checkpoints/drug effects , Vorinostat/pharmacology , Vorinostat/administration & dosage , Cell Proliferation/drug effects , Cyclin D1/genetics , Cyclin D1/metabolism
2.
Cancer Res ; 84(10): 1597-1612, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38588411

ABSTRACT

Resistance to immune checkpoint blockade (ICB) therapy represents a formidable clinical challenge limiting the efficacy of immunotherapy. In particular, prostate cancer poses a challenge for ICB therapy due to its immunosuppressive features. A ketogenic diet (KD) has been reported to enhance response to ICB therapy in some other cancer models. However, adverse effects associated with continuous KD were also observed, demanding better mechanistic understanding and optimized regimens for using KD as an immunotherapy sensitizer. In this study, we established a series of ICB-resistant prostate cancer cell lines and developed a highly effective strategy of combining anti-PD1 and anti-CTLA4 antibodies with histone deacetylase inhibitor (HDACi) vorinostat, a cyclic KD (CKD), or dietary supplementation of the ketone body ß-hydroxybutyrate (BHB), which is an endogenous HDACi. CKD and BHB supplementation each delayed prostate cancer tumor growth as monotherapy, and both BHB and adaptive immunity were required for the antitumor activity of CKD. Single-cell transcriptomic and proteomic profiling revealed that HDACi and ketogenesis enhanced ICB efficacy through both cancer cell-intrinsic mechanisms, including upregulation of MHC class I molecules, and -extrinsic mechanisms, such as CD8+ T-cell chemoattraction, M1/M2 macrophage rebalancing, monocyte differentiation toward antigen-presenting cells, and diminished neutrophil infiltration. Overall, these findings illuminate a potential clinical path of using HDACi and optimized KD regimens to enhance ICB therapy for prostate cancer. SIGNIFICANCE: Optimized cyclic ketogenic diet and 1,3-butanediol supplementation regimens enhance the efficacy of immune checkpoint blockade in prostate cancer through epigenetic and immune modulations, providing dietary interventions to sensitize tumors to immunotherapy.


Subject(s)
Diet, Ketogenic , Drug Resistance, Neoplasm , Epigenesis, Genetic , Immune Checkpoint Inhibitors , Prostatic Neoplasms , Male , Diet, Ketogenic/methods , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/immunology , Prostatic Neoplasms/diet therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Mice , Epigenesis, Genetic/drug effects , Animals , Cell Line, Tumor , Vorinostat/pharmacology , Vorinostat/therapeutic use , Vorinostat/administration & dosage , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , 3-Hydroxybutyric Acid , Xenograft Model Antitumor Assays , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors
3.
J Pediatr Hematol Oncol ; 44(1): e255-e259, 2022 01 01.
Article in English | MEDLINE | ID: mdl-33448719

ABSTRACT

Embryonal tumor with multilayered rosettes is a rare and highly malignant early childhood brain tumor. We report a case of embryonal tumor with multilayered rosettes in the parietooccipital region of a 2-year-old girl. Histopathology of the tumor demonstrated amplification of the 19q13.42 locus and strong positivity for LIN28A. Treatment was multimodal and included 3 surgical resections, adjuvant chemotherapy with autologous stem cell rescue, and focal radiotherapy. The use of the agents vorinostat and isotretinoin, and the addition of focal radiation have not been extensively described in this patient population, but may attribute to our patient's sustained remission at 2.5-years follow-up.


Subject(s)
Brain Neoplasms , Chromosomes, Human, Pair 19/genetics , Genetic Loci , Isotretinoin/administration & dosage , Neoplasms, Germ Cell and Embryonal , Stem Cell Transplantation , Vorinostat/administration & dosage , Autografts , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Chemoradiotherapy, Adjuvant , Child, Preschool , Female , Humans , Neoplasms, Germ Cell and Embryonal/genetics , Neoplasms, Germ Cell and Embryonal/pathology , Neoplasms, Germ Cell and Embryonal/therapy
4.
AIDS ; 36(1): 75-82, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34586085

ABSTRACT

OBJECTIVE: The aim of this study was to examine whether administering both vorinostat and disulfiram to people with HIV (PWH) on antiretroviral therapy (ART) is well tolerated and can enhance HIV latency reversal. DESIGN: Vorinostat and disulfiram can increase HIV transcription in PWH on ART. Together, these agents may lead to significant HIV latency reversal. METHODS: Virologically suppressed PWH on ART received disulfiram 2000 mg daily for 28 days and vorinostat 400 mg daily on days 8-10 and 22-24. The primary endpoint was plasma HIV RNA on day 11 relative to baseline using a single copy assay. Assessments included cell-associated unspliced RNA as a marker of latency reversal, HIV DNA in CD4+ T-cells, plasma HIV RNA, and plasma concentrations of ART, vorinostat, and disulfiram. RESULTS: The first two participants (P1 and P2) experienced grade 3 neurotoxicity leading to trial suspension. After 24 days, P1 presented with confusion, lethargy, and ataxia having stopped disulfiram and ART. Symptoms resolved by day 29. After 11 days, P2 presented with paranoia, emotional lability, lethargy, ataxia, and study drugs were ceased. Symptoms resolved by day 23. CA-US RNA increased by 1.4-fold and 1.3-fold for P1 and P2 respectively. Plasma HIV RNA was detectable from day 8 to 37 (peak 81 copies ml-1) for P2 but was not increased in P1 Antiretroviral levels were therapeutic and neuronal injury markers were elevated in P1. CONCLUSION: The combination of prolonged high-dose disulfiram and vorinostat was not safe in PWH on ART and should not be pursued despite evidence of latency reversal.


Subject(s)
HIV Infections , Disulfiram/administration & dosage , Drug Therapy, Combination/adverse effects , HIV Infections/drug therapy , Humans , Virus Latency/physiology , Vorinostat/administration & dosage
5.
Drug Deliv ; 28(1): 2301-2309, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34730060

ABSTRACT

Radiotherapy (RT) is a frequently used means in clinical tumor treatment. The outcome of RT varies, however, to a great extent, due to RT resistance or intolerable dose, which might be resolved by the development of radio-sensitizing strategies. Here, we report redox-sensitive iodinated polymersomes (RIP) carrying histone deacetylase inhibitor, suberoylanilide hydroxamic acid (SAHA, vorinostat), as a new dual-functional nano-radiosensitizer for breast cancer radiotherapy. SAHA-loaded RIP (RIP-SAHA) with a size of about 101 nm exhibited good colloidal stability while the reduction-activated release of SAHA, giving rise to better antitumor effect to 4T1 breast carcinoma cells than free SAHA. Accordingly, RIP-SAHA combined with a 4 Gy dose of X-ray radiation led to significantly enhanced suppression of 4T1 cells compared with SAHA combined 4 Gy of X-ray radiation, as a result of enhanced DNA damage and impeded DNA damage repair. The pharmacokinetics and biodistribution studies by single-photon emission computed tomography (SPECT) with 125I-labeled SAHA (125I-SAHA) showed a 17.3-fold longer circulation and 237.7-fold better tumor accumulation of RIP-SAHA over SAHA. The systemic administration of RIP-SAHA greatly sensitized radiotherapy of subcutaneous 4T1 breast tumors and brought about significant inhibition of tumor growth, without causing damages to major organs, compared with radiotherapy alone. RIP not only enhanced SAHA delivery but also acted as a radiosensitizer. RIP-SAHA emerges as a smart dual-functional nano-radiosensitizer to effectively enhance tumor radiotherapy.


Subject(s)
Breast Neoplasms/pathology , Histone Deacetylase Inhibitors/administration & dosage , Oxidation-Reduction/radiation effects , Radiation-Sensitizing Agents/administration & dosage , Vorinostat/administration & dosage , Animals , Cell Line, Tumor , Cell Proliferation/radiation effects , Cell Survival/radiation effects , Chemistry, Pharmaceutical , DNA Damage/radiation effects , Drug Carriers/chemistry , Drug Liberation , Female , Histone Deacetylase Inhibitors/pharmacokinetics , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Polymers/chemistry , Radiation-Sensitizing Agents/pharmacokinetics , Tissue Distribution , Vorinostat/pharmacokinetics
6.
Neurosci Lett ; 764: 136241, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34509565

ABSTRACT

Autism spectrum disorder (ASD) and intellectual disability (ID) are neurodevelopmental diseases associated with various gene mutations. Previous genetic and clinical studies reported that ASH1L is a high ASD risk gene identified in human patients. Our recent study used a mouse model to demonstrate that loss of ASH1L in the developing mouse brain was sufficient to cause multiple developmental defects, core autistic-like behaviors, and impaired cognitive memory, suggesting that the disruptive ASH1L mutations are the causative drivers leading the human ASD/ID genesis. Using this Ash1L-deletion-induced ASD/ID mouse model, here we showed that postnatal administration of vorinostat (SAHA), a histone deacetylase inhibitor (HDACi), significantly ameliorated both ASD-like behaviors and ID-like cognitive memory deficit. Thus, our study demonstrates that SAHA is a promising reagent for the pharmacological treatment of core ASD/ID behavioral and memory deficits caused by disruptive ASH1L mutations.


Subject(s)
Autism Spectrum Disorder/drug therapy , Cognitive Dysfunction/drug therapy , Histone Deacetylase Inhibitors/administration & dosage , Intellectual Disability/drug therapy , Vorinostat/administration & dosage , Animals , Autism Spectrum Disorder/genetics , Cognitive Dysfunction/genetics , DNA-Binding Proteins/genetics , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Histone-Lysine N-Methyltransferase/genetics , Humans , Intellectual Disability/genetics , Male , Memory/drug effects , Mice, Knockout , Social Skills
7.
Naunyn Schmiedebergs Arch Pharmacol ; 394(11): 2245-2257, 2021 11.
Article in English | MEDLINE | ID: mdl-34415354

ABSTRACT

Hepatocellular carcinoma (HCC) is a highly fatal form of liver cancer. Recently, the interest in using amino acids as therapeutic agents has noticeably grown. The present work aimed to evaluate the possible antiproliferative effects of selected amino acids supplementation or deprivation in human HCC cell lines and to investigate their effects on critical signaling molecules in HCC pathogenesis and the outcomes of their combination with the histone deacetylase inhibitor vorinostat. HepG2 and Huh7 cells were treated with different concentrations of L-leucine, L-glutamine, or L-methionine and cell viability was determined using MTT assay. Insulin-like growth factor 1 (IGF1), phosphorylated ribosomal protein S6 kinase (p70 S6K), p53, and cyclin D1 (CD1) protein levels were assayed using ELISA. Caspase-3 activity was assessed colorimetrically. L-leucine supplementation (0.8-102.4 mM) and L-glutamine supplementation (4-128 mM) showed dose-dependent antiproliferative effects in both cell lines but L-methionine supplementation (0.2-25.6 mM) only affected the viability of HepG2 cells. Glutamine or methionine deprivation suppressed the proliferation of HepG2 cells whereas leucine deprivation had no effect on cell viability in both cell lines. The combination between the effective antiproliferative changes in L-leucine, L-glutamine, and L-methionine concentrations greatly suppressed cell viability and increased the sensitivity to vorinostat in both cell lines. The growth inhibitory effects were paralleled with significant decreases in IGF-1, phospho p70 S6k, and CD1 levels and significant elevations in p53 and caspase-3 activity. Changes in amino acids concentrations could profoundly affect growth in HCC cell lines and their response to epigenetic therapy.


Subject(s)
Amino Acids/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Vorinostat/pharmacology , Amino Acids/administration & dosage , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/genetics , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Epigenesis, Genetic , Hep G2 Cells , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/pharmacology , Humans , Liver Neoplasms/genetics , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism , Vorinostat/administration & dosage
8.
J Clin Oncol ; 39(31): 3506-3514, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34270348

ABSTRACT

PURPOSE: 131I-metaiodobenzylguanidine (MIBG) is an active radiotherapeutic for neuroblastoma. The primary aim of this trial was to identify which of three MIBG regimens was likely associated with the highest true response rate. PATIENTS AND METHODS: Patients 1-30 years were eligible if they had relapsed or refractory neuroblastoma, at least one MIBG-avid site, and adequate autologous stem cells. Patients received MIBG 18 mCi/kg on day 1 and autologous stem cell on day 15. Patients randomly assigned to arm A received only MIBG; patients randomly assigned to arm B received intravenous vincristine on day 0 and irinotecan daily on days 0-4; patients randomly assigned to arm C received vorinostat (180 mg/m2/dose) orally once daily on days 1 to 12. The primary end point was response after one course by New Approaches to Neuroblastoma Therapy criteria. The trial was designed with 105 patients to ensure an 80% chance that the arm with highest response rate was selected. RESULTS: One hundred fourteen patients were enrolled, with three ineligible and six unevaluable, leaving 105 eligible and evaluable patients (36 in arm A, 35 in arm B, and 34 in arm C; 55 boys; and median age 6.5 years). After one course, the response rates (partial response or better) on arms A, B, and C were 14% (95% CI, 5 to 30), 14% (5 to 31), and 32% (18 to 51). An additional five, five, and four patients met New Approaches to Neuroblastoma Therapy Minor Response criteria on arms A, B, and C, respectively. On arms A, B, and C, rates of any grade 3+ nonhematologic toxicity after first course were 19%, 49%, and 35%. CONCLUSION: Vorinostat and MIBG is likely the arm with the highest true response rate, with manageable toxicity. Vincristine and irinotecan do not appear to improve the response rate to MIBG and are associated with increased toxicity.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Resistance, Neoplasm , Neoplasm Recurrence, Local/drug therapy , Neuroblastoma/drug therapy , Salvage Therapy , 3-Iodobenzylguanidine/administration & dosage , Adolescent , Adult , Child , Child, Preschool , Female , Follow-Up Studies , Humans , Infant , Irinotecan/administration & dosage , Male , Neoplasm Recurrence, Local/pathology , Neuroblastoma/pathology , Prognosis , Prospective Studies , Survival Rate , Vincristine/administration & dosage , Vorinostat/administration & dosage , Young Adult
9.
EBioMedicine ; 69: 103457, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34224975

ABSTRACT

BACKGROUND: Histone acetylation/deacetylase process is one of the most studied epigenetic modifications. Histone deacetylase inhibitors (HDACis) have shown clinical benefits in haematological malignancies but failed in solid tumours due to the lack of biomarker-driven stratification. METHODS: We perform integrative pharmaco-transcriptomic analysis by correlating drug response profiles of five pan-HDACis with transcriptomes of solid cancer cell lines (n=659) to systematically identify generalizable gene signatures associated with HDACis sensitivity and resistance. The established signatures are then applied to identify cancer subtypes that are potentially sensitive or resistant to HDACis, and drugs that enhance the efficacy of HDACis. Finally, the reproductivity of the established HDACis signatures is evaluated by multiple independent drug response datasets and experimental assays. FINDINGS: We successfully delineate generalizable gene signatures predicting sensitivity (containing 46 genes) and resistance (containing 53 genes) to all five HDACis, with their reproductivity confirmed by multiple external sources and independent internal assays. Using the gene signatures, we identify low-grade glioma harbouring isocitrate dehydrogenase 1/2 (IDH1/2) mutation and non-YAP1-driven subsets of small-cell lung cancer (SCLC) that particularly benefit from HDACis monotherapy. Further, based on the resistance gene signature, we identify clinically-approved Dasatinib as a synthetic lethal drug with HDACi, synergizing in inducing apoptosis and reactive oxygen species on a panel of SCLC. Finally, Dasatinib significantly enhances the therapeutic efficacy of Vorinostat in SCLC xenografts. INTERPRETATION: Our work establishes robust gene signatures predicting HDACis sensitivity/resistance in solid cancer and uncovers combined Dasatinib/HDACi as a synthetic lethal combination therapy for SCLC. FUNDING: This work was supported by the National Natural Science Foundation of China (82072570 to F. Yao; 82002941 to B. Sun).


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Dasatinib/therapeutic use , Drug Resistance, Neoplasm , Histone Deacetylase Inhibitors/therapeutic use , Lung Neoplasms/genetics , Transcriptome , Adaptor Proteins, Signal Transducing/genetics , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Dasatinib/administration & dosage , Drug Synergism , Humans , Isocitrate Dehydrogenase/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice, Inbred NOD , Mice, SCID , Mutation , Pharmacogenomic Variants , Transcription Factors/genetics , Vorinostat/administration & dosage , Vorinostat/therapeutic use , YAP-Signaling Proteins
10.
Pathol Oncol Res ; 27: 636088, 2021.
Article in English | MEDLINE | ID: mdl-34257602

ABSTRACT

Objective: Uterine carcinosarcoma (UCS) is a rare but highly aggressive malignancy with biphasic growth pattern. This morphology can be attributed to epithelial-mesenchymal transition (EMT) that often associates with tumor invasion and metastasis. Accordingly, we analyzed a novel patient-derived preclinical model to explore whether EMT is a potential target in UCS. Methods: A novel UCS cell line (PF338) was established from the malignant pleural effusion of a 59-year-old patient at time of disease progression. Immunohistochemistry was performed in primary and metastatic tumor lesions. Oncogenic mutations were identified by next-generation sequencing. Viability assays and cell cycle analyses were used to test in vitro sensitivity to different standard and novel treatments. E-cadherin, ß-catenin and pSMAD2 expressions were measured by immunoblot. Results: Whereas immunohistochemistry of the metastatic tumor showed a predominantly sarcomatous vimentin positive tumor that has lost E-cadherin expression, PF338 cells demonstrated biphasic growth and carried mutations in KRAS, PIK3CA, PTEN and ARID1A. PF338 tumor cells were resistant to MEK- and TGF-ß signaling-inhibition but sensitive to PIK3CA- and PARP-inhibition and first-line chemotherapeutics. Strikingly, histone deacetylase (HDAC) inhibition markedly reduced cell viability by inducing a dose-dependent G0/1 arrest and led to mesenchymal-epithelial transition as evidenced by morphological change and increased E-cadherin and ß-catenin expression. Conclusions: Our data suggest that HDAC inhibition is effective in a novel UCS cell line by interfering with both viability and differentiation. These findings emphasize the dynamic manner of EMT/MET and epigenetics and the importance of molecular profiling to pave the way for novel therapies in UCS.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinosarcoma/pathology , Cell Cycle Checkpoints , Epithelial-Mesenchymal Transition , Histone Deacetylases/chemistry , Pleural Effusion, Malignant/pathology , Uterine Neoplasms/pathology , Biomarkers, Tumor/genetics , Carcinosarcoma/drug therapy , Carcinosarcoma/metabolism , Cisplatin/administration & dosage , Female , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Mutation , Paclitaxel/administration & dosage , Phthalazines/administration & dosage , Piperazines/administration & dosage , Pleural Effusion, Malignant/drug therapy , Pleural Effusion, Malignant/metabolism , Prognosis , Pyrazoles/administration & dosage , Quinolines/administration & dosage , Tumor Cells, Cultured , Uterine Neoplasms/drug therapy , Uterine Neoplasms/metabolism , Vorinostat/administration & dosage
11.
Cancer Biother Radiopharm ; 36(8): 632-641, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34252288

ABSTRACT

Background: Histone deacetylase (HDAC) inhibitors have been shown in preclinical studies to upregulate norepinephrine transporters in neuroblastoma and pheochromocytoma, and somatostatin receptors in pulmonary carcinoid, small cell lung cancer, and pancreatic neuroendocrine malignancies. This pilot imaging study in humans focuses on midgut neuroendocrine carcinoma metastatic to the liver, evaluating the effect of pretreatment with the HDAC inhibitor vorinostat on uptake of 123I-MIBG and 68Ga-DOTATOC. Materials and Methods: Multiple midgut neuroendocrine liver metastases in clinically stable subjects were imaged with 123I-MIBG and 68Ga-DOTATOC before and after a 4-d course of vorinostat. Scans were performed with strict attention to detail and timed about 1 month apart occurring just before monthly long-acting octreotide administrations. Uptake changes in tumor and normal liver parenchyma were assessed on positron emission computed tomography (PET/CT) with standardized uptake values and on single photon emission computed tomography (SPECT) with qualitative ratio images. Results: The experimental units were metastatic liver lesions within patients (n = 50). There was no significant difference in administered activity or uptake time between pairs of scans for either radiotracer. Statistically significant increase in maximum standardized uptake values (SUVmax) averaged over all lesions was noted on the 68Ga-DOTATOC PET scans (+11%, p < 0.01). SUVmax in normal liver showed no significant change (p = 0.12). There was no qualitative change in uptake of 123I-MIBG after vorinostat. Conclusions: In this pilot imaging study in patients with midgut neuroendocrine liver metastases, a short course of the HDAC inhibitor vorinostat induced a statistically significant increase in SUVmax on 68Ga-DOTATOC PET/computed tomography (CT) imaging in some hepatic neuroendocrine tumor metastases. There was no significant effect of vorinostat on tumor uptake of 123I-MIBG on SPECT/CT imaging. Given the pilot nature of this trial, the findings merit further investigation with a more rigorous protocol evaluating longer pretreatment and different dosages of vorinostat or other HDAC inhibitors, as well as effects on the therapeutic capability of 177Lu- or 90Y-somatostatin analogs.


Subject(s)
3-Iodobenzylguanidine/pharmacology , Intestinal Neoplasms , Liver Neoplasms , Neoplasm Metastasis/diagnostic imaging , Neuroendocrine Tumors , Octreotide/analogs & derivatives , Pancreatic Neoplasms , Single Photon Emission Computed Tomography Computed Tomography/methods , Stomach Neoplasms , Vorinostat , Biological Availability , Female , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/pharmacokinetics , Humans , Intestinal Neoplasms/drug therapy , Intestinal Neoplasms/pathology , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Male , Middle Aged , Multimodal Imaging/methods , Neoplasm Staging , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/pathology , Octreotide/pharmacology , Outcome Assessment, Health Care , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Pilot Projects , Radiopharmaceuticals/pharmacology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Vorinostat/administration & dosage , Vorinostat/pharmacokinetics
12.
Pharmacol Res ; 171: 105787, 2021 09.
Article in English | MEDLINE | ID: mdl-34314859

ABSTRACT

We adopted a novel strategy by combining histone deacetylase (HDAC) inhibitors with traditional chemotherapeutics to treat solid tumors. However, chemotherapeutics often have a narrow therapeutic index and need multiple administrations with undesired side effects that lead to the intolerance. To reduce the non-specificity of chemotherapeutics, targeted therapy was introduced to restrict such agents in the tumor with minimum effects on other tissues. We developed bioinspired artificial exosomes (AE), which enabled to deliver chemotherapeutics to the tumors effectively after systemic administration. AE were produced by incorporating membrane proteins from cancer cells into phospholipid liposomes that mimicked the plasma membrane. The synthesized AE were used for the delivery of broad-spectrum chemotherapeutic doxorubicin (DOX) and vorinostat (SAHA), an epigenetic inhibitor. The combination of DOX and SAHA showed synergistic effects on suppressing non-small cell lung cancer cells and xenograft tumors without apparent adverse effects. AE facilitated the delivery of drugs to tumor tissue and extended the retention time of drugs within tumors. Taken together, these studies suggest that the bioengineered artificial exosomes may serve as novel delivery strategy for chemotherapeutics to treat non-small cell lung cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Doxorubicin/administration & dosage , Drug Carriers/administration & dosage , Exosomes , Histone Deacetylase Inhibitors/administration & dosage , Lung Neoplasms/drug therapy , Vorinostat/administration & dosage , Animals , Antineoplastic Agents/chemistry , Cell Line , Doxorubicin/chemistry , Drug Carriers/chemistry , Drug Liberation , Epigenesis, Genetic , Humans , Lung Neoplasms/pathology , Mice, Inbred BALB C , Tumor Burden/drug effects , Vorinostat/chemistry
13.
Drug Deliv ; 28(1): 963-972, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34036867

ABSTRACT

PD-1/PD-L1 blocking therapy has become one of the most promising methods in the field of tumor treatment. However, it encounters the challenge of immune escape due to the exhaustion of T cells. Studies have shown that the epigenetic regulation drug histone deacetylase inhibitor (HDACi) may be able to reverse exhausted T cells by changing the epigenetic transcription program. Therefore, the combination of epigenetic therapy and PD-1/PD-L1 blockade therapy is expected to reverse the immune escape, whereas the overriding goal should aim at the spontaneous release and synergy of PD-1/PD-L1 blocking siRNA and HDACi. In this study, we develop PDDS{polyethylene glycol-b-asparaginate(diethylenetriamine-vorinostat), (PEG-b-P[Asp(DET-SAHA)n] PPDS)}encapsulating siRNA-PD-L1to provide micelles siRNA-PD-L1-loaded micelles (siRNA@PPDS). Transmission electron microscope (TEM) images demonstrate that siRNA@PPDS micelles presented spherical morphology with a size of about 120 nm; hydrodynamic data analysis indicates pH sensitivity of siRNA@PPDS micelles. The experiments reveal that siRNA@PPDS micelles could be well uptaken by the tumor cells to silence the expression of PD-L1 protein in a dose-dependent manner; compared with the free SAHA, the SAHA-loaded micelles PPDS show higher cytotoxicity to induce tumor cell apoptosis and block cell cycle in G1 phase on melanoma-bearing mice, siRNA@PPDS has shown outstanding inhibition of tumor growth and pulmonary metastasis. By comprehensively activating the immune system, lysosome activable polymeric vorinostat encapsulating PD-L1KD for the combination therapy of PD-L1-KD and HDACIs can be an effective strategy to reverse the unresponsiveness of immune checkpoint inhibitors and a promising treatment to inhibit tumor growth, recurrence, and metastasis in clinic.


Subject(s)
Antineoplastic Agents/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Micelles , Programmed Cell Death 1 Ligand 2 Protein/antagonists & inhibitors , RNA, Small Interfering/pharmacology , Vorinostat/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Survival , Chemistry, Pharmaceutical , Dose-Response Relationship, Drug , Drug Carriers/chemistry , Drug Liberation , Epigenesis, Genetic , G1 Phase/drug effects , Histone Deacetylase Inhibitors/administration & dosage , Hydrogen-Ion Concentration , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Particle Size , Polyethylene Glycols/chemistry , RNA, Small Interfering/administration & dosage , Vorinostat/administration & dosage , Xenograft Model Antitumor Assays
14.
Cell Death Dis ; 12(6): 532, 2021 05 24.
Article in English | MEDLINE | ID: mdl-34031358

ABSTRACT

Human intestinal peptide transporter PEPT1 is commonly repressed in human colorectal cancer (CRC), yet its relationship with sensitivity to the common CRC treatment ubenimex has not previously been elucidated. In this study, we confirmed PEPT1 suppression in CRC using real-time quantitative polymerase chain reaction and western blotting and then investigated the underlying epigenetic pathways involved using bisulfite sequencing, chromatin immunoprecipitation, siRNA knockdown, and reporter gene assays. We found that PEPT1 transcriptional repression was due to both DNMT1-mediated DNA methylation of the proximal promoter region and HDAC1-mediated histone deacetylation, which blocked P300-mediated H3K18/27Ac at the PEPT1 distal promoter. Finally, the effects of the epigenetic activation of PEPT1 on the CRC response to ubenimex were evaluated using sequential combination therapy of decitabine and ubenimex both in vitro and in xenografts. In conclusion, epigenetic silencing of PEPT1 due to increased DNMT1 and HDAC1 expression plays a vital role in the poor response of CRC to ubenimex.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Histone Deacetylase Inhibitors/pharmacology , Peptide Transporter 1/genetics , Animals , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Methylation/drug effects , Drug Synergism , Epigenesis, Genetic/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors/administration & dosage , Humans , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/pharmacology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Leucine/administration & dosage , Leucine/analogs & derivatives , Mice , Mice, Inbred BALB C , Mice, Nude , Peptide Transporter 1/metabolism , Vorinostat/administration & dosage , Vorinostat/pharmacology , Xenograft Model Antitumor Assays
15.
Cancer Lett ; 508: 76-91, 2021 06 28.
Article in English | MEDLINE | ID: mdl-33775711

ABSTRACT

The development of a new generation of tyrosine kinase inhibitors (TKIs) has improved the treatment response in lung adenocarcinomas. However, acquired resistance often occurs due to new epidermal growth factor receptor (EGFR) mutations. In particular, the C797S mutation confers drug resistance to T790M-targeting EGFR TKIs. To address C797S resistance, a promising therapeutic avenue is combination therapy that targets both total EGFR and acquired mutations to increase drug efficacy. We showed that combining vorinostat, a histone deacetylase inhibitor (HDACi), with brigatinib, a TKI, enhanced antitumor effects in primary culture and cell lines of lung adenocarcinomas harboring EGFR L858R/T790M/C797S mutations (EGFR-3M). While EGFR phosphorylation was decreased by brigatinib, vorinostat reduced total EGFR-3M (L858R/T790M/C797S) proteins through STUB1-mediated ubiquitination and degradation. STUB1 preferably ubiquitinated other EGFR mutants and facilitated protein turnover compared to EGFR-WT. The association between EGFR and STUB1 required the functional chaperone-binding domain of STUB1 and was further enhanced by vorinostat. Finally, STUB1 levels modulated EGFR downstream functions. Low STUB1 expression was associated with significantly poorer overall survival than high STUB1 expression in patients harboring mutant EGFR. Vorinostat combined with brigatinib significantly improved EGFR-TKI sensitivity to EGFR C797S by inducing EGFR-dependent cell death and may be a promising therapy in treating C797S-resistant lung adenocarcinomas.


Subject(s)
Adenocarcinoma of Lung/drug therapy , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Lung Neoplasms/drug therapy , Organophosphorus Compounds/pharmacology , Pyrimidines/pharmacology , Vorinostat/pharmacology , Adenocarcinoma of Lung/enzymology , Adenocarcinoma of Lung/genetics , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm , Drug Synergism , ErbB Receptors/genetics , HEK293 Cells , Humans , Lung Neoplasms/enzymology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation , Organophosphorus Compounds/administration & dosage , Pyrimidines/administration & dosage , Random Allocation , Vorinostat/administration & dosage , Xenograft Model Antitumor Assays
16.
Clin Lymphoma Myeloma Leuk ; 21(3): 154-161.e3, 2021 03.
Article in English | MEDLINE | ID: mdl-33478922

ABSTRACT

INTRODUCTION: Outcomes continue to improve in relapsed myeloma as more effective treatment options emerge. We report a multicenter single-arm phase 2 trial evaluating toxicity and efficacy of the histone deacetylase (HDAC) inhibitor vorinostat in combination with bortezomib and dexamethasone. PATIENTS AND METHODS: Sixteen patients who had received a median of 1 prior treatment line received bortezomib subcutaneously 1.3 mg/m2 days 1, 4, 8, and 11; dexamethasone 20 mg orally days 1-2, 4-5, 8-9, and 11-12; vorinostat 400 mg orally days 1-4, 8-11, and 15-18 of a 21-day cycle. After receipt of a minimum of 3 cycles of therapy, participants received maintenance vorinostat (400 mg days 1-4 and 15-18 of a 28-day cycle). RESULTS: Overall response was 81.3%: complete response occurred in 4 of 16, very good partial response in 2 of 16, and partial response 7 of 16. Clinical benefit response rate was 100%; median progression-free survival was 11.9 months. A total of 75% patients experienced a dose reduction or stopped treatment as a result of intolerability. CONCLUSION: Although toxicity and dose reductions were observed, this study demonstrates that the combination of vorinostat, bortezomib, and dexamethasone is effective in relapsed myeloma with good response rates, suggesting there is an ongoing rationale for further optimization of HDAC inhibitor-based combinations in the treatment of myeloma to improve tolerability and enhance efficacy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Multiple Myeloma/drug therapy , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biomarkers, Tumor , Bortezomib/administration & dosage , Dexamethasone/administration & dosage , Female , Histone Deacetylase Inhibitors/administration & dosage , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Molecular Targeted Therapy , Multiple Myeloma/diagnosis , Multiple Myeloma/etiology , Multiple Myeloma/mortality , Neoplasm Staging , Prognosis , Recurrence , Retreatment , Treatment Outcome , Vorinostat/administration & dosage
17.
Neurotox Res ; 39(2): 170-181, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33400178

ABSTRACT

Suberoylanilide hydroxamic acid (SAHA/Vorinostat), a potent inhibitor of histone deacetylases (HDACs), is known to possess antidepressant properties. However, the exact mechanisms underlying this activity are unknown. In this study, we evaluated the effect of SAHA on the expression of GluN2A, GluN2B (NMDA receptor subunits), (p-)AMPK, and ΔFos proteins which are an integral part of the signal transduction pathways in the brain and also involved in the pathophysiology of depression as well as the mechanism of antidepressant action. We also measured the concentration of malondialdehyde (MDA - a product of lipid peroxidation). The study was carried out in the prefrontal cortex (PFC) and hippocampus (Hp), brain regions implicated in depression. Although SAHA induced changes in the expression of all the proteins and MDA concentration, the effects differed depending on the drug dose, time, and brain structure involved. SAHA reduced MDA concentration and significantly increased p-AMPK protein expression, indicating it may prevent oxidative stress. SAHA also increased the levels of HDAC3 and NMDA subunits (GluN2A and GluN2B), implying it is neuroprotective and may play a crucial role in synaptic plasticity. Moreover, ΔFosB and FosB levels were significantly elevated, suggesting that SAHA may modulate learning and memory processes. Overall, the data indicate that the Hp might play a pivotal role in the mechanism of action of SAHA, hinting at novel mechanisms it play in the antidepressant and neuroprotective effects of SAHA.


Subject(s)
Antidepressive Agents/administration & dosage , Hippocampus/drug effects , Histone Deacetylase Inhibitors/administration & dosage , Oxidative Stress/drug effects , Prefrontal Cortex/drug effects , Signal Transduction/drug effects , Vorinostat/administration & dosage , Animals , Hippocampus/metabolism , Male , Mice, Inbred C57BL , Prefrontal Cortex/metabolism
18.
Invest New Drugs ; 39(3): 812-820, 2021 06.
Article in English | MEDLINE | ID: mdl-33409898

ABSTRACT

BACKGROUND: Until the advent of T cell check point inhibitors standard second-line therapy for patients with metastatic urothelial cancer (mUC) was undefined. Histone deacetylase inhibitors (HDACi) have anti-cancer activity in a variety of tumor models including modulation of apoptosis in bladder cancer cell lines. We evaluated the efficacy and toxicity of the HDACi vorinostat in patients with mUC failing first-line platinum-based therapy either in the adjuvant/neoadjuvant setting or for recurrent/advanced disease. METHODS: Vorinostat was given orally 200 mg twice daily continuously until progression or unacceptable toxicity. The primary end point was RECIST response rate (RR); a RR > 20% was deemed interesting in a 2-stage design requiring one response in the first 12 patients to proceed to 2nd stage for a total of 37 subjects. CT or MRI scan imaging occurred every 6 weeks. RESULTS: Fourteen patients were accrued characterized by: median age 66 years (43-84); Caucasian (79%); males (86%); and Karnofsky performance status ≥90 (50%). Accrual was terminated in the first stage as no responses were observed. Best response was stable disease (3 patients). Progression was observed in 8 patients. Two patients came off therapy prior to re-imaging and a 3rd patient died while on treatment and was not assessed for response. Median number of cycles was 2 (range 1-11). Median disease-free survival and overall survival times were 1.1 (0.8, 2.1) & 3.2 (2.1, 14.5) months, respectively. Toxicities were predominantly cytopenias and thrombocytopenic bleeding. Two pts. had grade 5 toxicity unlikely related to treatment. Two pts. had grade 4 and 6 had grade 3 toxicities observed. Two patients with stable disease remained on therapy for 6+ cycles. CONCLUSIONS: Vorinostat on this dose-schedule had limited efficacy and significant toxicity resulting in a unfavorable risk:benefit ratio in patients with mUC. NCT00363883.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Transitional Cell/drug therapy , Histone Deacetylase Inhibitors/administration & dosage , Neoplasm Recurrence, Local/drug therapy , Urologic Neoplasms/drug therapy , Vorinostat/administration & dosage , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Carcinoma, Transitional Cell/mortality , Carcinoma, Transitional Cell/pathology , Female , Histone Deacetylase Inhibitors/adverse effects , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Treatment Outcome , Urologic Neoplasms/mortality , Urologic Neoplasms/pathology , Urothelium/pathology , Vorinostat/adverse effects
20.
Clin Lymphoma Myeloma Leuk ; 21(2): 97-105, 2021 02.
Article in English | MEDLINE | ID: mdl-33158772

ABSTRACT

BACKGROUND: Sézary syndrome (SS) and mycosis fungoides (MF), 2 types of cutaneous T-cell lymphoma, cause significant morbidity and adversely affect patients' quality of life (QoL). The present study assessed the QoL measurement changes in patients receiving mogamulizumab versus vorinostat. PATIENTS AND METHODS: A multicenter phase III trial was conducted of patients with stage IB-IV MF/SS with ≥ 1 failed systemic therapy. The QoL measures included Skindex-29 and the Functional Assessment of Cancer Therapy-General. The symptoms, function, and QoL subdomains were longitudinally modeled using mixed models with prespecified covariates. Meaningful change thresholds (MCTs) were defined using distribution-based methods. The categorical changes by group over time and the time to clinically meaningful worsening were analyzed. RESULTS: Of the 372 randomized patients, mogamulizumab demonstrated improvement in Skindex-29 symptoms (cycles 3, 5, and 7; P < .05) and functional (cycles 3 and 5; P < .05) scales. A significantly greater proportion of mogamulizumab-treated patients improved by MCTs or more from baseline in the Skindex-29 symptoms domain (cycles 3, 5, 7, and 11) and functioning domain (cycle 5). Significant differences in the Functional Assessment of Cancer Therapy-General physical well-being (cycles 1, 3, and 5; P < .05) were observed in favor of mogamulizumab and a greater proportion of patients had declined by MCTs or more at cycles 1, 3, 5, and 7 with vorinostat treatment. The median time to symptom worsening using Skindex-29 was 27.4 months for mogamulizumab versus 6.6 months for vorinostat. In the patients with SS, the time to worsening favored mogamulizumab (P < .005) for all Skindex-29 domains. The time to worsening was similar for the 2 MF treatment arms. CONCLUSION: The symptoms, function, and overall QoL of patients with MF/SS favored mogamulizumab over vorinostat across all time points. Patients with the greatest symptom burden and functional impairment derived the most QoL benefit from mogamulizumab.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Mycosis Fungoides/drug therapy , Quality of Life , Sezary Syndrome/drug therapy , Skin Neoplasms/drug therapy , Vorinostat/administration & dosage , Aged , Female , Humans , Male , Middle Aged , Minimal Clinically Important Difference , Mycosis Fungoides/complications , Mycosis Fungoides/psychology , Neoplasm Staging , Receptors, CCR4/antagonists & inhibitors , Sezary Syndrome/complications , Sezary Syndrome/psychology , Skin Neoplasms/complications , Skin Neoplasms/psychology , Time Factors , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...