Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Sci Rep ; 10(1): 18058, 2020 10 22.
Article in English | MEDLINE | ID: mdl-33093534

ABSTRACT

Fragile X syndrome (FXS), an X-chromosome linked intellectual disability, is the leading monogenetic cause of autism spectrum disorder (ASD), a neurodevelopmental condition that currently has no specific drug treatment. Building upon the demonstrated therapeutic effects on spatial memory of bryostatin-1, a relatively specific activator of protein kinase C (PKC)ε, (also of PKCα) on impaired synaptic plasticity/maturation and spatial learning and memory in FXS mice, we investigated whether bryostatin-1 might affect the autistic phenotypes and other behaviors, including open field activity, activities of daily living (nesting and marble burying), at the effective therapeutic dose for spatial memory deficits. Further evaluation included other non-spatial learning and memory tasks. Interestingly, a short period of treatment (5 weeks) only produced very limited or no therapeutic effects on the autistic and cognitive phenotypes in the Fmr1 KO2 mice, while a longer treatment (13 weeks) with the same dose of bryostatin-1 effectively rescued the autistic and non-spatial learning deficit cognitive phenotypes. It is possible that longer-term treatment would result in further improvement in these fragile X phenotypes. This effect is clearly different from other treatment strategies tested to date, in that the drug shows little acute effect, but strong long-term effects. It also shows no evidence of tolerance, which has been a problem with other drug classes (mGluR5 antagonists, GABA-A and -B agonists). The results strongly suggest that, at appropriate dosing and therapeutic period, chronic bryostatin-1 may have great therapeutic value for both ASD and FXS.


Subject(s)
Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/therapy , Bryostatins/administration & dosage , Bryostatins/physiology , Cognition Disorders/genetics , Cognition Disorders/therapy , Fragile X Syndrome/genetics , Fragile X Syndrome/therapy , Animals , Behavior, Animal , Bryostatins/pharmacology , Learning , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Protein Kinase C/metabolism , Spatial Memory
2.
J Alzheimers Dis ; 67(2): 555-570, 2019.
Article in English | MEDLINE | ID: mdl-30530975

ABSTRACT

BACKGROUND: Bryostatin-activated PKC epsilon pre-clinically induces synaptogenesis, anti-apoptosis, anti-amyloid-ß oligomers, and anti-hyperphosphorylated tau. OBJECTIVES: To investigate bryostatin safety, tolerability, and efficacy to improve cognition in advanced Alzheimer's disease (AD) patients. METHODS: A double-blind, randomized, placebo-controlled Phase II, 12-week trial of i.v. bryostatin for 150 advanced AD patients (55-85) with MMSE-2 of 4-15, randomized 1:1:1 into 20 µg and 40 µg bryostatin, and placebo arms. The Full Analysis Set (FAS) and the Completer Analysis Set (CAS) were pre-specified alternative assessments (1-sided, p < 0.1 for primary efficacy, and 2-sided, p < 0.05 for pre-specified and post hoc exploratory analyses). RESULTS: The safety profile was similar for 20 µg treatment and placebo patients. The 40 µg patients showed safety and drop-out issues, but no efficacy. Primary improvement of Severe Impairment Battery (SIB) scores at 13 weeks was not significant (p = 0.134) in the FAS, although in the CAS, the SIB comparison favored 20 µg bryostatin compared to placebo patients (p < 0.07). Secondary analyses at weeks 5 and 15 (i.e., 30 days post-final dosing) also favored 20 µg bryostatin compared to placebo patients. A pre-specified ANCOVA for baseline memantine blocking bryostatin and positive post-hoc trend analyses were statistically significant (2-sided, p < 0.05). CONCLUSION: Although the primary endpoint was not significant in the FAS, primary and secondary analyses in the CAS, and pre-specified and post-hoc exploratory analyses did favor bryostatin 20 µg compared to the placebo cohort. These promising Phase II results support further trials of 20 µg bryostatin- without memantine- to treat AD.


Subject(s)
Alzheimer Disease/drug therapy , Bryostatins/adverse effects , Bryostatins/therapeutic use , Activities of Daily Living , Aged , Aged, 80 and over , Alzheimer Disease/psychology , Bryostatins/administration & dosage , Cognition , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Infusions, Intravenous , Male , Mental Status and Dementia Tests , Middle Aged , Negative Results , Patient Dropouts , Treatment Outcome
3.
Acta Parasitol ; 63(1): 198-209, 2018 Mar 26.
Article in English | MEDLINE | ID: mdl-29351079

ABSTRACT

Accumulating evidence suggest that some infectious agents may interfere in the natural progression of neoplasia. This study examined the association between chronic infection with adult Syphacia muris parasites and 1,2-dimethylhydrazine (DMH)-induced colorectal carcinogenesis in rats. In addition, the conceivable therapeutic effect of Bryostatin-1, a potent extract of the marine Bryozoan, Bugulane ritina, was investigated against this combined effect.DMH administration has induced aberrant crypt foci (ACF), surrogate biomarkers for colorectal carcinogenesis, while the S. muris infection combined with DMH has significantly increased the total numbers of ACF. Nonetheless, treatment with Bryostatin-1 after infection has significantly reduced the ACF numbers particularly larger ones. This inhibition was concomitant with significant inhibition in the immunohistochemical levels of the ki67, Caspase-3 and IgM levels in colorectal epithelium, as well as serum levels of IgM and IgG. Additionally, treatment with Bryostatin-1 after S. muris + DMH has modulated enzymatic antioxidative markers levels of superoxide dismutase and catalase as well as the non-enzymatic antioxidant markers levels of reduced glutathione, lipid peroxidation, nitric oxide and total antioxidant capacity. Further, treatment with Bryostatin-1 has down-regulated the mRNA expression levels of COX-2 and APC genes in colorectal mucosa. In conclusion, infection with S. muris during colorectal carcinogenesis has significantly modulated the oxidative stress markers in the colorectum, while treatment with Bryostatin-1 has exerted significant curative potential. A mechanism could be explained that Bryostatin-1 treatment has reduced oxidative stress markers activities along with affecting host to parasite immunity possibly leading to changes in the COX-2 and APC expression, retarding cellular proliferation and subsequently reducing the colorectal carcinogenesis events.


Subject(s)
Antineoplastic Agents/administration & dosage , Bryostatins/administration & dosage , Carcinogenesis , Colorectal Neoplasms/physiopathology , Oxidative Stress , Oxyuriasis/parasitology , Oxyuroidea/physiology , Animals , Biomarkers, Tumor/analysis , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/pathology , Disease Models, Animal , Gene Expression , Immunohistochemistry , Oxyuriasis/complications , Rats , Treatment Outcome
4.
AIDS ; 30(9): 1385-92, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26891037

ABSTRACT

OBJECTIVE: The protein kinase C (PKC) agonist bryostatin-1 has shown significant ex-vivo potency to revert HIV-1 latency, compared with other latency reversing agents (LRA). The safety of this candidate LRA remains to be proven in treated HIV-1-infected patients. METHODS: In this pilot, double-blind phase I clinical-trial (NCT 02269605), we included aviraemic HIV-1-infected patients on triple antiretroviral therapy to evaluate the effects of two different single doses of bryostatin-1 (10 or 20 µg/m) compared with placebo. RESULTS: Twelve patients were included, four in each arm. Bryostatin-1 was well tolerated in all participants. Two patients in the 20 µg/m arm developed grade 1 headache and myalgia. No detectable increases of cell-associated unspliced (CA-US) HIV-1-RNA were observed in any study arm, nor differences in HIV-1 mRNA dynamics between arms (P = 0.44). The frequency of samples with low-level viraemia did not differ between arms and low-level viraemia did not correlate with CA-US HIV-1-RNA levels (P = 0.676). No changes were detected on protein kinase C (PKC) activity and in biomarkers of inflammation (sCD14 and interleukin-6) in any study arm. After the single dose of bryostatin-1, plasma concentrations were under detection limits in all the patients in the 10 µg/m arm, and below 50 pg/ml (0.05 nmol/l) in those in the 20 µg/m arm. CONCLUSION: Bryostatin-1 was safe at the single doses administered. However, the drug did not show any effect on PKC activity or on the transcription of latent HIV, probably due to low plasma concentrations. This study will inform next trials aimed at assessing higher doses, multiple dosing schedules or combination studies with synergistic drugs.


Subject(s)
Anti-Retroviral Agents/therapeutic use , Bryostatins/administration & dosage , Bryostatins/adverse effects , Enzyme Activators/administration & dosage , Enzyme Activators/adverse effects , HIV Infections/drug therapy , Virus Latency/drug effects , Adult , Double-Blind Method , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/pathology , Female , Humans , Male , Middle Aged , Pilot Projects , Placebos/administration & dosage , Treatment Outcome
5.
Eur J Pharmacol ; 764: 404-412, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26189021

ABSTRACT

Blood-brain barrier (BBB) disruption and hemorrhagic transformation (HT) following ischemic/reperfusion injury contributes to post-stroke morbidity and mortality. Bryostatin, a potent protein kinase C (PKC) modulator, has shown promise in treating neurological injury. In the present study, we tested the hypothesis that administration of bryostatin would reduce BBB disruption and HT following acute ischemic stroke; thus, prolonging the time window for administering recombinant tissue plasminogen activator (r-tPA). Acute cerebral ischemia was produced by reversible occlusion of the right middle cerebral artery (MCAO) in 18-20-month-old female rats using an autologous blood clot with delayed r-tPA reperfusion. Bryostatin (or vehicle) was administered at 2 h post-MCAO and r-tPA was administered at 6 h post-MCAO. Functional assessment, lesion volume, and hemispheric swelling measurements were performed at 24 h post-MCAO. Assessment of BBB permeability, measurement of hemoglobin, assessment of matrix metalloproteinase (MMP) levels by gel zymography, and measurement of PKCε, PKCα, PKCδ expression by western blot were conducted at 24 h post-MCAO. Rats treated with bryostatin prior to r-tPA administration had decreased mortality and hemispheric swelling when compared with rats treated with r-tPA alone. Administration of bryostatin also limited BBB disruption and HT and down-regulated MMP-9 expression while up-regulating PKCε expression at 24 h post-MCAO. Bryostatin administration ameliorates BBB disruption and reduces the risk of HT by down-regulating MMP-9 activation and up-regulating PKCε. In this proof-of-concept study, bryostatin treatment lengthened the time-to-treatment window and enhanced the efficacy and safety of thrombolytic therapy.


Subject(s)
Blood-Brain Barrier/drug effects , Bryostatins/administration & dosage , Fibrinolytic Agents/administration & dosage , Infarction, Middle Cerebral Artery/drug therapy , Intracranial Hemorrhages/prevention & control , Neuroprotective Agents/administration & dosage , Thrombolytic Therapy , Time-to-Treatment , Tissue Plasminogen Activator/administration & dosage , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Capillary Permeability/drug effects , Disease Models, Animal , Drug Administration Schedule , Female , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Intracranial Hemorrhages/metabolism , Intracranial Hemorrhages/pathology , Matrix Metalloproteinase 9/metabolism , Protein Kinase C-epsilon/metabolism , Rats, Sprague-Dawley , Time Factors , Up-Regulation
6.
J Pharmacol Exp Ther ; 349(3): 393-401, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24659806

ABSTRACT

Fragile X syndrome (FXS) is caused by transcriptional silencing in neurons of the FMR1 gene product, fragile X mental retardation protein (FMRP), a repressor of dendritic mRNA translation. The lack of FMRP leads to dysregulation of synaptically driven protein synthesis and impairments of intellect, cognition, and behavior, a disorder that currently has no effective therapeutics. Fragile X mice were treated with chronic bryostatin-1, a relatively selective protein kinase ε activator with pharmacological profiles of rapid mGluR desensitization, synaptogenesis, and synaptic maturation/repairing. Differences in the major FXS phenotypes, synapses, and cognitive functions were evaluated and compared among the age-matched groups. Long-term treatment with bryostatin-1 rescues adult fragile X mice from the disorder phenotypes, including normalization of most FXS abnormalities in hippocampal brain-derived neurotrophic factor expression and secretion, postsynaptic density-95 levels, glycogen synthase kinase-3ß phosphorylation, transformation of immature dendritic spines to mature synapses, densities of the presynaptic and postsynaptic membranes, and spatial learning and memory. Our results show that synaptic and cognitive function of adult FXS mice can be normalized through pharmacologic treatment and that bryostatin-1-like agents may represent a novel class of drugs to treat fragile X mental retardation even after postpartum brain development has largely completed.


Subject(s)
Bryostatins/therapeutic use , Fragile X Syndrome/drug therapy , Hippocampus/drug effects , Maze Learning/drug effects , Memory/drug effects , Spatial Behavior/drug effects , Synapses/drug effects , Animals , Bryostatins/administration & dosage , Dendritic Spines/drug effects , Dendritic Spines/pathology , Disease Models, Animal , Fragile X Syndrome/metabolism , Fragile X Syndrome/pathology , Fragile X Syndrome/physiopathology , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Male , Mice , Mice, Inbred Strains , Microscopy, Confocal , Microscopy, Electron , Protein Kinase C-epsilon/metabolism , Synapses/metabolism , Synapses/physiology , Synapses/ultrastructure
7.
Invest New Drugs ; 30(2): 723-8, 2012 Apr.
Article in English | MEDLINE | ID: mdl-20936324

ABSTRACT

BACKGROUND: The California Cancer Consortium has performed a Phase II trial of infusional bryostatin, a protein kinase C inhibitor isolated from the marine invertebrate bryozoan, Bugula Neritina, a member of the phylum Ectoprocta, in combination with cisplatin, in patients (pts) with recurrent platinum-sensitive or resistant ovarian cancer (OC). METHODS: Pts received bryostatin 45 mcg/m(2) as a 72 h continuous infusion followed by cisplatin 50 mg/m(2). Cycles were repeated every 3 weeks. Dosages were chosen based on phase I data obtained by the CCC in a population of pts with mixed tumor types. RESULTS: Eight pts with recurrent or persistent epithelial OC received 23 cycles of treatment. All pts had received previous platinum-based chemotherapy; two pts had received one prior course, five had received two prior courses, and one had received three prior courses of chemotherapy. The median age was 64 (range 32-72), and Karnofsky performance status 90 (range 80-100). A median of 3 cycles of chemotherapy were delivered (range: 1-5). The median progression-free and overall survivals were 3 and 8.2 months respectively. Best responses included two partial responses (one in a platinum-resistant pt), three pts with stable disease, and three progressions. All pts experienced Grade 3 or 4 toxicities including severe myalgias/pain/fatigue/asthenia in six pts, and severe nausea/vomiting/constipation in two other pts. One pt experienced a seizure and liver function tests were elevated in one other. CONCLUSIONS: A modest response rate is observed in pts with recurrent or persistent ovarian cancer treated with the combination of bryostatin and cisplatin. The toxicity profile, however, observed in this pt population (primarily severe myalgias), precludes tolerability and prevents this combination from further investigation at this dose and schedule. It is possible that platinum pre-exposure in OC patients exacerbates observed toxicity. Phase II dosages of investigational agents in OC pts that are determined by phase I trials in pts with other tumor types should be chosen cautiously.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasm Recurrence, Local , Neoplasms, Glandular and Epithelial/drug therapy , Ovarian Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bryostatins/administration & dosage , California , Carcinoma, Ovarian Epithelial , Cisplatin/administration & dosage , Drug Administration Schedule , Female , Humans , Infusions, Intravenous , Kaplan-Meier Estimate , Middle Aged , Neoplasms, Glandular and Epithelial/enzymology , Neoplasms, Glandular and Epithelial/mortality , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Kinase Inhibitors/administration & dosage , Time Factors , Treatment Outcome
8.
Leuk Res ; 35(1): 87-94, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20598742

ABSTRACT

PURPOSE: Pharmacologic differentiating agents have had relatively limited clinical success outside of the use of ATRA in acute promyelocytic leukemia and DNA methyltransferase inhibitors in myelodysplastic syndromes. The differentiating effects of such agents can be enhanced in combination with lineage-specific growth factors. We developed a dose finding trial to assess toxicity, differentiating activity, and clinical impact of the combination of bryostatin-1 and GM-CSF. EXPERIMENTAL DESIGN: Patients with poor risk myeloid malignancies were eligible to enroll in a dose finding study of continuous infusion bryostatin-1 combined with a fixed dose of daily GM-CSF. Toxicities were graded per NCI CTC version 2.0 and pharmacokinetic and correlative study samples were obtained to assess the combination's clinical and biologic differentiating effects. RESULTS: Thirty-two patients were treated with the combination therapy and the dose determined to be most suitable for study in a larger trial was continuous infusion broystatin-1 at 16µg/m(2) for 14 days and subcutaneous GM-CSF at 125µg/m(2) daily for 14 days every 28 days. Arthralgias and myalgias limited further dose escalation. Clinically, the combination impacted differentiation with improvement of absolute neutrophil counts (p=0.0001) in the majority of patients. Interestingly, there were two objective clinical responses, including a CR after a single cycle. Both the bryostatin-1 plasma concentrations and the correlative studies supported biologic activity of the combination at the doses where clinical responses were observed. CONCLUSIONS: Combining growth factors with pharmacologic differentiating agents may increase their clinical effectiveness and further studies should focus on such combinations.


Subject(s)
Antineoplastic Agents/administration & dosage , Bryostatins/administration & dosage , Cell Differentiation , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Adult , Aged , Antineoplastic Agents/therapeutic use , Bryostatins/therapeutic use , Dose-Response Relationship, Drug , Female , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Humans , Male , Middle Aged , Prognosis
9.
Article in English | MEDLINE | ID: mdl-20540089

ABSTRACT

BACKGROUND: Bryostatin-1, a highly oxygenated marine macrolide with a unique polyacetate backbone isolated from the marine animal Bugula neritina (Linnaeus), is now being developed as an anti-cancer drug for treating malignancy. In the present study, developmental toxicity of bryostatin-1 was evaluated in Sprague-Dawley rats. METHODS: Bryostatin-1 was intravenously administered to rats on gestation days 6-15 at 4.0, 8.0, and 16.0 microg/kg on a daily basis. Then the reproductive parameters were determined in animals, and fetuses were examined for external, visceral, and skeletal malformations. RESULTS: The total weight gains were significantly different in animals between the control group and 8.0 and 16.0 microg/kg bryostatin-1 groups during and after treatment. The resorption and death fetus rates were significantly different between the bryostatin-1 group (16 microg/kg) and the control group. The fetal weight and fetal crown-rump length in the bryostatin-1 groups were significantly lower than that in the control group. CONCLUSIONS: Our results indicated that maternal toxicity occurred when the dose of bryostatin-1 was at 8.0 microg/kg, embryotoxicity at 16.0 microg/kg, and fetotoxicity at 4.0 microg/kg; but bryostatin-1 showed no teratogenic effect in rats. In light of our findings, bryostatin-1 should be used with caution in pregnant women with cancer, if they would like to continue the pregnancy.


Subject(s)
Bryostatins/toxicity , Embryonic Development/drug effects , Fetus/drug effects , Toxicity Tests , Animals , Bryostatins/administration & dosage , Bryostatins/chemistry , Female , Male , Maternal Exposure , Pregnancy , Rats , Rats, Sprague-Dawley , Teratogens/toxicity
10.
Am J Clin Oncol ; 33(2): 121-4, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19738452

ABSTRACT

PURPOSE: To determine the efficacy and toxicity of the protein kinase C inhibitor bryostatin-1 plus paclitaxel in patients with advanced pancreatic carcinoma. METHODS: Each treatment cycle consisted of paclitaxel 90 mg/m by intravenous infusion over 1 hour on days 1, 8, and 16, plus bryostatin 25 mcg/m as a 1-hour intravenous infusion on days 2, 9, and 15, given every 28 days. Patients were evaluated for response after every 2 treatment cycles, and continued therapy until disease progression or prohibitive toxicity. The primary objective was to determine whether the combination produced a response rate of at least 30%. RESULTS: Nineteen patients with locally advanced or metastatic pancreatic adenocarcinoma received a total of 52 cycles of therapy (range: 1-10). Patients received the combination as first-line therapy for advanced disease (N = 5) or after prior chemotherapy used alone or in combination with local therapy. No patients had a confirmed objective response. The median time to treatment failure was 1.9 months (95% confidence intervals: 1.2, 2.6 months). Reasons for discontinuing therapy included progressive disease or death in 14 patients (74%) or because of adverse events or patient choice in 5 patients (26%). The most common grade 3 to 4 toxicities included leukopenia in 26%, anemia in 11%, myalgias in 11%, gastrointestinal bleeding in 11%, infection in 10%, and thrombosis in 10%. CONCLUSION: The combination of weekly paclitaxel and bryostatin-1 is not an effective therapy for patients with advanced pancreatic carcinoma.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Pancreatic Neoplasms/drug therapy , Protein Kinase C/antagonists & inhibitors , Adenocarcinoma/secondary , Adult , Aged , Aged, 80 and over , Bryostatins/administration & dosage , Female , Humans , Male , Middle Aged , Neoplasm Staging , Paclitaxel/administration & dosage , Pancreatic Neoplasms/pathology , Survival Rate , Treatment Outcome , Young Adult
11.
Am J Hematol ; 84(8): 484-7, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19536846

ABSTRACT

Bryostatin 1, isolated from a marine bryozoan, enhances the efficacy of cytotoxic agents through modulation of the protein kinase C pathway and is active in combination with vincristine for diffuse large B-cell lymphoma. Further, the apoptotic frequency of peripheral blood T lymphocytes as determined by flow cytometry may predict which patients will respond to this combination. We tested the efficacy and safety of bryostatin 1 50 microg/m(2) given over 24 hr and vincristine 1.4 mg/m(2) on days 1 and 15 every 28 days in aggressive B-cell non-Hodgkin lymphoma (NHL) relapsing after autologous stem cell transplantation. End points included tumor response, toxicity, and survival. Responses were correlated with an increase in apoptotic frequency of CD5+ cells by flow cytometry using annexin V staining. Fourteen patients were enrolled with 13 being evaluable for a response. The overall response rate was 31% with two patients achieving a complete response. The most common toxicities were Grade 3 lymphopenia (seven patients), Grade 3 to 4 neutropenia (two patients), and Grade 3 hypophosphatemia (two patients). Median progression-free and overall survivals for all patients were 5.7 and 21.4 months, respectively. One patient demonstrated an increase in T-cell apoptotic frequency, also achieving a complete response. Bryostatin 1 and vincristine have efficacy in select patients with aggressive NHL. Future investigations of agents targeting the protein kinase C pathway may benefit from early response assessment using flow cytometry to evaluate T-cell apoptosis.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Lymphoma, B-Cell/prevention & control , Stem Cell Transplantation , Aged , Annexin A5/metabolism , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Apoptosis/drug effects , Bryostatins/administration & dosage , Bryostatins/adverse effects , CD5 Antigens/blood , Disease-Free Survival , Female , Humans , Lymphoma, B-Cell/blood , Lymphopenia/blood , Lymphopenia/chemically induced , Male , Middle Aged , Protein Kinase C/metabolism , Recurrence , Survival Rate , T-Lymphocytes/metabolism , Transplantation, Autologous , Vincristine/administration & dosage , Vincristine/adverse effects
12.
Behav Pharmacol ; 19(3): 245-56, 2008 May.
Article in English | MEDLINE | ID: mdl-18469542

ABSTRACT

Previous work demonstrated that protein kinase C (PKC) is implicated in learning and memory. This study investigated whether: (i) PKC activated by bryostatin-1 (Bryo) just before or just after sessions of classical conditioning was capable of enhancing classical conditioning of the rabbit nictitating membrane response; (ii) improved behavioral performance matched the time course of PKC activation induced by Bryo; and (iii) vitamin E (Vit E) enhanced the efficacy of Bryo. Paired rabbits received daily trace conditioning with a tone conditioned stimulus and a corneal air puff unconditioned stimulus. Unpaired rabbits received the same stimuli but in an explicitly unpaired manner. After trace conditioning, all rabbits received daily delay conditioning, and then tone intensity testing. Rabbits pretreated with 10 microg/kg Bryo every other day before a relatively simple trace conditioning task showed more conditioned responses (CRs) during the first 10 trials of each trace conditioning session and a higher likelihood of a CR on the first trial of each trace conditioning session than rabbits pretreated with the vehicle control. Rabbits either posttreated daily with 10 microg/kg Bryo or pretreated with Vit E and subjected to a difficult trace conditioning task showed increased CRs relative to the vehicle control. Neither Bryo nor Vit E or their combination altered nonassociative responding or altered sensitivity to the conditioned stimulus or unconditioned stimulus. These findings demonstrate Bryo has long-term enhancing effects on classical conditioning of the rabbit nictitating membrane response.


Subject(s)
Antineoplastic Agents/pharmacology , Bryostatins/pharmacology , Conditioning, Classical/drug effects , Protein Kinase C/drug effects , Animals , Antineoplastic Agents/administration & dosage , Bryostatins/administration & dosage , Cognition/drug effects , Drug Synergism , Male , Nictitating Membrane/drug effects , Nictitating Membrane/metabolism , Protein Kinase C/metabolism , Rabbits , Vitamin E/pharmacology , Vitamins/pharmacology
13.
Pancreas ; 36(4): 346-52, 2008 May.
Article in English | MEDLINE | ID: mdl-18437080

ABSTRACT

OBJECTIVES: Protein kinase C (PKC) is involved in tumor growth and apoptosis and hence represents a potential target for cancer therapy. This study investigated the expression of PKC in pancreatic tumor tissue in comparison to adjacent normal tissue and determined the modulation of PKC by bryostatin-1 (BRYO) on pancreatic cancer cell lines. METHODS: Pancreatic tissue was obtained from 18 patients who had a resection (14 with ductal adenocarcinoma and 4 with adenoma and high-grade dysplasia). Cytosolic and nuclear membrane PKCs in the paired samples were determined by immunoblotting. HPAC cells were treated with gemcitabine and BRYO and in sequential and concomitant combination. To evaluate cell viability, apoptosis, and electrophoretic mobility shift assay, 3-(4,5-dimetylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, enzyme-linked immunosorbent assay, and nuclear factor kappaB (NF-kappaB) assays were used. RESULTS: As compared with the adjacent normal tissue, PKC-alpha, PKC-beta1, and PKC-delta were higher in the tumor; PKC-epsilon was higher in the normal tissue. Pretreatment with gemcitabine followed by BRYO resulted in decreased cell viability, increased apoptosis, and inhibited NF-kappaB than either agent alone or BRYO followed by gemcitabine. CONCLUSION: Protein kinase C is overexpressed and activated in pancreatic cancer as compared with normal tissue. Inhibition of PKC could sensitize pancreatic cancer cell lines to the effects of gemcitabine. The potentiation of gemcitabine by BRYO is sequence-dependent and mediated through inhibition of PKC-dependent activation of NF-kappaB.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Isoenzymes/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/enzymology , Protein Kinase C/antagonists & inhibitors , Adenocarcinoma/drug therapy , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Adenoma/drug therapy , Adenoma/enzymology , Adenoma/pathology , Bryostatins/administration & dosage , Cell Survival/drug effects , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Humans , Isoenzymes/metabolism , Pancreatic Neoplasms/pathology , Protein Kinase C/metabolism , Protein Kinase Inhibitors/therapeutic use , Reference Values , Gemcitabine
14.
Eur J Pharmacol ; 584(2-3): 328-37, 2008 Apr 28.
Article in English | MEDLINE | ID: mdl-18313045

ABSTRACT

Evidence is emerging that protein kinase C (PKC) plays a crucial role in the neural processing of memory information and that PKC deficits underlie certain types of memory impairment, including Alzheimer's dementia. Chronic activation of PKC isozymes with bryostatin-1 induces synthesis of the proteins that are involved in memory consolidation and, therefore, may represent a pharmacological strategy for antidementic and memory therapies. PKC isozymes are, however, sensitive to oxidants, whose generation is also increased by PKC activation. Oxidants may be responsible for some adverse effects with PKC activators, potentially limiting their antidementic and memory-enhancing "benefit". We investigated the effects of intravenous bryostatin-1, a potent PKC activator, and of its co-administration with oral alpha-tocopherol, a potent antioxidant, on spatial learning and memory. Bryostatin-1 at a chronic and intravenous dose of 10 microg/m2 (2 doses/week for 3 weeks) alone did not significantly affect the spatial learning and memory, but showed a synergistic effect when co-administered with alpha-tocopherol (60 IU/kg, orally and daily for 3 weeks), a potent lipid-soluble antioxidant and also a possible inhibitor of PKC in peripheral tissues. Acute administration of the same doses, however, did not have obvious influence on the learning and memory. These results provide support for the strategy of achieving memory-enhancing benefits with PKC activators and restricting their oxidant-related adverse effects with alpha-tocopherol co-administration. These agents, therefore, may hold significant potential as new, combined antidementic and memory therapeutics in the future.


Subject(s)
Antioxidants/pharmacology , Behavior, Animal/drug effects , Bryostatins/pharmacology , Enzyme Activators/pharmacology , Learning/drug effects , Memory/drug effects , Nootropic Agents/pharmacology , alpha-Tocopherol/pharmacology , Administration, Oral , Animals , Antioxidants/administration & dosage , Brain/drug effects , Brain/enzymology , Bryostatins/administration & dosage , Dose-Response Relationship, Drug , Drug Synergism , Enzyme Activation , Enzyme Activators/administration & dosage , Injections, Intravenous , Male , Maze Learning/drug effects , Nootropic Agents/administration & dosage , Protein Kinase C/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , alpha-Tocopherol/administration & dosage
15.
Cancer Chemother Pharmacol ; 62(5): 875-80, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18270704

ABSTRACT

BACKGROUND: We sought to determine the response rate and toxicity profile of sequential paclitaxel and bryostatin-1, a novel, selective inhibitor of protein kinase C, in patients with advanced esophageal cancer. PATIENTS AND METHODS: Patients with advanced esophageal and gastroesophageal junction cancer were enrolled. All gave informed consent. They were initially treated with paclitaxel 90 mg/m(2) intravenously on Day 1 and bryostatin-1 50 microg/m2 on Day 2 weekly for three consecutive weeks out of four. Because of severe myalgias, dosing was reduced to paclitaxel 80 mg/m2 with bryostatin-1 40 microg/m2 and then to paclitaxel 80 mg/m2 with bryostatin-1 25 microg/m2. RESULTS: Twenty-four patients were enrolled, with 22 assessable for response. The partial response rate was 27%. 10 patients treated with bryostatin-1 40-50 microg/m2 had a response rate of 40 versus 17% at bryostatin-1 25 microg/m2 (p-value = 0.3). Median time-to-progression was 3.7 months and median survival was 8.3 months. Grade 3/4 myalgias were seen in 50% of patients. Myalgias appeared to be related to bryostatin-1 dose. Because of toxicity, the trial was closed prior to full accrual. CONCLUSIONS: Despite potential anti-tumor activity of this combination in patients with advanced esophageal cancer, further development is not warranted, given the severe toxicity, especially myalgias, that were seen.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Esophageal Neoplasms/drug therapy , Adenocarcinoma/pathology , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bryostatins/administration & dosage , Carcinoma, Squamous Cell/pathology , Disease Progression , Esophageal Neoplasms/pathology , Female , Humans , Male , Middle Aged , Paclitaxel/administration & dosage
16.
Acad Radiol ; 14(12): 1531-9, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18035282

ABSTRACT

RATIONALE AND OBJECTIVES: A reliable noninvasive method for in vivo detection of early therapeutic response of non-Hodgkin's lymphoma (NHL) patients would be of great clinical value. This study evaluates the feasibility of (1)H and (31)P magnetic resonance spectroscopy (MRS) for in vivo detection of response to combination chemotherapy of human diffuse large B-cell lymphoma (DLCL2) xenografts in severe combined immunodeficient (SCID) mice. MATERIALS AND METHODS: Combination chemotherapy with cyclophosphamide, hydroxy doxorubicin, Oncovin, prednisone, and bryostatin 1 (CHOPB) was administered to tumor-bearing SCID mice weekly for up to four cycles. Spectroscopic studies were performed before the initiation of treatment and after each cycle of the CHOPB. Proton MRS for detection of lactate and total choline was performed using a selective multiple-quantum-coherence-transfer (Sel-MQC) and a spin-echo-enhanced Sel-MQC (SEE-Sel-MQC) pulse sequence, respectively. Phosphorus-31 MRS using a nonlocalized, single-pulse sequence without proton decoupling was also performed on these animals. RESULTS: Significant decreases in lactate and total choline were detected in the DLCL2 tumors after one cycle of CHOPB chemotherapy. The ratio of phosphomonoesters to beta-nucleoside triphosphate (PME/betaNTP, measured by (31)P MRS) significantly decreased in the CHOPB-treated tumors after two cycles of CHOPB. The control tumors did not exhibit any significant changes in either of these metabolites. CONCLUSIONS: This study demonstrates that (1)H and (31)P MRS can detect in vivo therapeutic response of NHL tumors and that lactate and choline offer a number of advantages over PMEs as markers of early therapeutic response.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Lymphoma, B-Cell/drug therapy , Lymphoma, Large B-Cell, Diffuse/drug therapy , Magnetic Resonance Spectroscopy/methods , Neoplasm Transplantation , Transplantation, Heterologous , Animals , Antibiotics, Antineoplastic/administration & dosage , Antineoplastic Agents/administration & dosage , Antineoplastic Agents, Alkylating , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Bryostatins/administration & dosage , Choline/analysis , Cyclophosphamide/administration & dosage , Doxorubicin/administration & dosage , Feasibility Studies , Glycerophosphates/analysis , Humans , Hydrogen , Lactates/analysis , Mice , Mice, SCID , Nucleotides/analysis , Phosphorus , Prednisone/administration & dosage , Treatment Outcome , Vincristine/administration & dosage
17.
Leuk Res ; 31(10): 1393-402, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17391757

ABSTRACT

Natural killer (NK) cells are potent effectors of innate antitumor defense and are currently exploited for immune-based therapy of human leukemia. However, malignant blood cells in acute myeloid leukemia (AML) display low levels of ligands for the activating immunoreceptor NKG2D and can thus evade NK immunosurveillance. We examined the possibility of up-regulating NKG2D-specific UL16-binding protein (ULBP) ligands using anti-neoplastic compounds with myeloid differentiation potential. Combinations of 5-aza-2'-deoxycytidine, trichostatin A, vitamin D3, bryostatin-1, and all-trans-retinoic acid, used together with myeloid growth factors and interferon-gamma, increased cell surface ULBP expression up to 10-fold in the AML cell line HL60 and in primary AML blasts. Up-regulation of ULBP ligands was associated with induction of myelomonocytic differentiation of AML cells. Higher ULBP expression increased NKG2D-dependent sensitivity of HL60 cells to NK-mediated killing. These findings identify NKG2D ligands as targets of leukemia differentiation therapy and suggest a clinical benefit in combining a pharmacological approach with NK cell-based immunotherapy in AML.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cytotoxicity, Immunologic , Intercellular Signaling Peptides and Proteins/metabolism , Killer Cells, Natural/immunology , Leukemia, Myeloid/metabolism , Acute Disease , Azacitidine/administration & dosage , Azacitidine/analogs & derivatives , Bryostatins/administration & dosage , Cell Differentiation , Cell Line, Tumor , Cholecalciferol/administration & dosage , Decitabine , Flow Cytometry , GPI-Linked Proteins , Humans , Hydroxamic Acids/administration & dosage , Leukemia, Myeloid/immunology , Leukemia, Myeloid/pathology , Reverse Transcriptase Polymerase Chain Reaction , Tretinoin/administration & dosage , Up-Regulation
18.
Clin Cancer Res ; 12(23): 7059-62, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17145828

ABSTRACT

PURPOSE: Bryostatin 1 is a macrocyclic lactone with protein kinase C inhibitory activity. Gemcitabine is a nucleotide analogue with a broad spectrum of anticancer activity. Bryostatin 1 enhanced the activity of antitumor agents including gemcitabine in preclinical models. The primary objective of this phase I study was to determine the recommended doses for phase II trials of bryostatin 1 and gemcitabine. EXPERIMENTAL DESIGN: Eligible patients had histologic or cytologic diagnosis of nonhematologic cancer refractory to conventional treatment; life expectancy of >3 months; normal renal, hepatic, and bone marrow function; and a Southwest Oncology Group performance status of 0 to 2. Gemcitabine was administered i.v. over 30 minutes and was followed by bryostatin 1 by i.v. infusion over 24 hours on days 1, 8, and 15 of a 28-day cycle. Bryostatin 1 (microg/m(2)) and gemcitabine (mg/m(2)) doses were escalated as follows: 25/600, 25/800, 25/1,000, 30/1,000, 35/1,000, and 45/1,000, respectively. RESULTS: Thirty-six patients (mean age, 57 years; male/female 15:21) were treated. The median number of treatment cycles per patient was 3 (range, 0-24). Four patients developed dose limiting toxicities: myalgia, 2; myelosuppression, 1; and elevation of serum alanine aminotransferase levels, 1. Ten grade 3 toxicities were observed (anemia, 2; neutropenia, 5; thrombocytopenia, 3). No treatment-related death was seen. The recommended doses for phase II trials for bryostatin 1 and gemcitabine were 35 microg/m(2) and 1,000 mg/m(2), respectively. Two heavily pretreated patients with breast and colon cancer experienced partial responses lasting 22 and 8 months, respectively. Eight patients had stable disease. CONCLUSION: The combination of bryostatin 1 and gemcitabine seemed to be well tolerated with limited grade 3 toxicity. The recommended dose of bryostatin 1 in combination with full doses of gemcitabine was 35 microg/m(2).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Bryostatins/administration & dosage , Deoxycytidine/analogs & derivatives , Neoplasms/drug therapy , Adolescent , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bryostatins/adverse effects , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Follow-Up Studies , Humans , Injections, Intravenous , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/diagnosis , Treatment Outcome , Gemcitabine
19.
Invest New Drugs ; 24(2): 141-9, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16514482

ABSTRACT

PURPOSE: Bryostatin-1 is a PKC modulator with direct anti-tumor activity and immunomodulatory properties. We combined different doses of Bryostatin-1 with IL-2 to determine effects on clinical response rate and T cell phenotype in patients with advanced kidney cancer. EXPERIMENTAL DESIGN: IL-2 naïve patients were given 11 x 10(6) IU subcutaneously of IL-2 on days 1-4, 8-11, and 15-18 of every 28-day cycle. Twenty four patients were randomized to treatment cohorts of 5, 15 or 25 mcg/m2 of Bryostatin-1 on days 1, 8 and 15, starting in the second cycle. An additional nine, non-randomized patients were given 35 mcg/m2. Lymphocytes were analyzed for number, activation status, and production of IL-2, IL-4 and IFN-gamma. Response evaluation was performed every 3 cycles. RESULTS: Common grade 3 toxicities included fatigue (5), nausea/vomiting (5), myopathy (3), dyspnea (3), and syncope (3). Four patients, in the two highest dose cohorts, demonstrated evidence of tumor shrinkage, although there was only 1 objective PR. The median time to progression was 104 days (95% CI 88-120) and the median survival was 452 days (95% CI = 424-480). There was no significant boosting effect of Bryostatin-1 on lymphocytes. CONCLUSIONS: The addition of Bryostatin-1 to IL-2 was well tolerated, but the overall response rate was low (3.2%), indicating that further studies with this combination are not warranted.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bryostatins/administration & dosage , Carcinoma, Renal Cell/drug therapy , Interleukin-2/administration & dosage , Kidney Neoplasms/drug therapy , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bryostatins/adverse effects , Carcinoma, Renal Cell/immunology , Cytokines/biosynthesis , Dose-Response Relationship, Drug , Female , Humans , Interleukin-2/adverse effects , Kidney Neoplasms/immunology , Male , Middle Aged , Neoplasm Metastasis , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Treatment Failure
SELECTION OF CITATIONS
SEARCH DETAIL
...