Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 313
Filter
1.
J Biol Chem ; 300(1): 105509, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38042493

ABSTRACT

Today, the majority of patients with pediatric B cell precursor acute lymphoblastic leukemia (BCP-ALL, hereafter ALL) survive their disease, but many of the survivors suffer from life-limiting late effects of the treatment. ALL develops in the bone marrow, where the cells are exposed to cAMP-generating prostaglandin E2. We have previously identified the cAMP signaling pathway as a putative target for improved efficacy of ALL treatment, based on the ability of cAMP signaling to reduce apoptosis induced by DNA damaging agents. In the present study, we have identified the antioxidant N-acetyl cysteine (NAC) as a powerful modifier of critical events downstream of the cell-permeable cAMP analog 8-(4-chlorophenylthio) adenosine-3', 5'- cyclic monophosphate (8-CPT). Accordingly, we found NAC to turn 8-CPT into a potent killer of ALL cells in vitro both in the presence and absence of DNA damaging treatment. Furthermore, we revealed that NAC in combination with 8-CPT is able to delay the progression of ALL in a xenograft model in NOD-scid IL2Rγnull mice. NAC was shown to rely on the ability of 8-CPT to activate the guanine-nucleotide exchange factor EPAC, and we demonstrated that the ALL cells are killed by apoptosis involving sustained elevated levels of calcium imposed by the combination of the two drugs. Taken together, we propose that 8-CPT in the presence of NAC might be utilized as a novel strategy for treating pediatric ALL patients, and that this powerful combination might be exploited to enhance the therapeutic index of current ALL targeting therapies.


Subject(s)
Acetylcysteine , Cyclic AMP , Guanine Nucleotide Exchange Factors , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Thionucleotides , Animals , Child , Humans , Mice , Acetylcysteine/pharmacology , Acetylcysteine/therapeutic use , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cyclic AMP/therapeutic use , DNA/drug effects , Guanine Nucleotide Exchange Factors/agonists , Mice, Inbred NOD , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Male , Female , Child, Preschool , Thionucleotides/pharmacology , Thionucleotides/therapeutic use , DNA Damage , Drug Therapy, Combination
2.
Neurobiol Dis ; 140: 104870, 2020 07.
Article in English | MEDLINE | ID: mdl-32294521

ABSTRACT

Spinal muscular atrophy (SMA) is a severe, inherited disease characterized by the progressive degeneration and death of motor neurons of the anterior horns of the spinal cord, which results in muscular atrophy and weakness of variable severity. Its early-onset form is invariably fatal in early childhood, while milder forms lead to permanent disability, physical deformities and respiratory complications. Recently, two novel revolutionary therapies, antisense oligonucleotides and gene therapy, have been approved, and might prove successful in making long-term survival of these patients likely. In this perspective, a deep understanding of the pathogenic mechanisms and of their impact on the interactions between motor neurons and other cell types within the central nervous system (CNS) is crucial. Studies using SMA animal and cellular models have taught us that the survival and functionality of motor neurons is highly dependent on a whole range of other cell types, namely glial cells, which are responsible for a variety of different functions, such as neuronal trophic support, synaptic remodeling, and immune surveillance. Thus, it emerges that SMA is likely a non-cell autonomous, multifactorial disease in which the interaction of different cell types and disease mechanisms leads to motor neurons failure and loss. This review will introduce the different glial cell types in the CNS and provide an overview of the role of glial cells in motor neuron degeneration in SMA. Furthermore, we will discuss the relevance of these findings so far and the potential impact on the success of available therapies and on the development of novel ones.


Subject(s)
Muscular Atrophy, Spinal/pathology , Neuroglia/pathology , Animals , Astrocytes/metabolism , Central Nervous System/metabolism , Disease Models, Animal , Genetic Therapy , Humans , Mice , Motor Neurons/pathology , Neuroglia/metabolism , Oligonucleotides, Antisense/therapeutic use , Spinal Cord/pathology , Survival of Motor Neuron 1 Protein/genetics , Thionucleotides/therapeutic use
3.
Mol Ther ; 25(5): 1069-1075, 2017 05 03.
Article in English | MEDLINE | ID: mdl-28366767

ABSTRACT

Oligonucleotides (oligos) have been under clinical development for approximately the past 30 years, beginning with antisense oligonucleotides (ASOs) and apatmers and followed about 15 years ago by siRNAs. During that lengthy period of time, numerous clinical trials have been performed and thousands of trial participants accrued onto studies. Of all the molecules evaluated as of January 2017, the regulatory authorities assessed that six provided clear clinical benefit in rigorously controlled trials. The story of these six is given in this review.


Subject(s)
Aptamers, Nucleotide/therapeutic use , Morpholinos/therapeutic use , Oligonucleotides, Antisense/therapeutic use , Oligonucleotides/therapeutic use , Polydeoxyribonucleotides/therapeutic use , Thionucleotides/therapeutic use , Clinical Trials as Topic , Cytomegalovirus Retinitis/genetics , Cytomegalovirus Retinitis/therapy , Cytomegalovirus Retinitis/virology , Drug Approval , Hepatic Veno-Occlusive Disease/genetics , Hepatic Veno-Occlusive Disease/pathology , Hepatic Veno-Occlusive Disease/therapy , Humans , Hypercholesterolemia/genetics , Hypercholesterolemia/pathology , Hypercholesterolemia/therapy , Macular Degeneration/genetics , Macular Degeneration/pathology , Macular Degeneration/therapy , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , Muscular Atrophy, Spinal/therapy , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/therapy
4.
Lancet Oncol ; 18(11): 1532-1542, 2017 11.
Article in English | MEDLINE | ID: mdl-29033099

ABSTRACT

BACKGROUND: Docetaxel and cabazitaxel improve overall survival compared with mitoxantrone in patients with metastatic castration-resistant prostate cancer. Custirsen (OGX011) is a second generation highly specific antisense oligonucleotide that inhibits the production of clusterin, an antiapoptotic protein that is upregulated in response to chemotherapy and that confers treatment resistance. We aimed to assess whether custirsen in combination with cabazitaxel and prednisone increases overall survival in patients with metastatic castration-resistant prostate cancer previously treated with docetaxel. METHODS: In this randomised, open-label, international, phase 3 trial, men with radiographically documented metastatic castration-resistant prostate cancer that had progressed after docetaxel treatment with a Karnofsky performance status of more than 70% and who were fit for chemotherapy, were recruited from 95 cancer treatment centres in eight countries. Patients were randomly assigned (1:1) centrally using permuted blocks (block size 8) to receive cabazitaxel plus prednisone (cabazitaxel 25 mg/m2 intravenously every 21 days plus oral prednisone 10 mg daily) with or without custirsen (640 mg intravenously on days 1, 8, and 15, plus three previous loading doses) until disease progression, unacceptable toxicity, or the completion of ten treatment cycles. Randomisation was stratified by use of opioids for prostate cancer-related pain at screening, disease progression following first-line docetaxel treatment established by radiographic evidence, and previous treatment with abiraterone or enzalutamide. The co-primary endpoints were overall survival in all randomly assigned patients and in a poor-prognosis subgroup. All analyses were intention to treat with the exception of safety, which was reported for patients who received any assigned treatment. The trial has been completed and the results presented here are the final analysis. This trial is registered with Clinicaltrials.gov, number NCT01578655. FINDINGS: Between Sept 9, 2012, and Sept 29, 2014, 795 patients were screened for enrolment. 635 men were eligible for inclusion and were randomly assigned (n=317 in the cabazitaxel and prednisone plus custirsen group and n=318 in the cabazitaxel and prednisone group). Median follow up was 28·3 months (IQR 24·4-34·5) for the custirsen group and 29·8 months (IQR 25·3-35·2) for the control group. Median overall survival in all randomly assigned patients did not differ between the two groups (14·1 months [95% CI 12·7-15·9] in the curtisen group vs 13·4 months [12·1-14·9] in the control group; hazard ratio [HR] 0·95 [95% CI 0·80-1·12]; log-rank p=0·53). In the poor prognosis subgroup, median overall survival also did not differ between the two treatment groups (11·0 months [95% CI 9·3-13·3] in the custursin group vs 10·9 months [8·2-12·4] in the control group; HR 0·97 [95% CI 0·80-1·21]; two-sided p=0·80). The most frequently reported grade 3 or worse adverse events in the custirsen versus control groups were neutropenia (70 [22%] of 315 vs 61 [20%] of 312), anaemia (68 [22%] vs 49 [16%]), fatigue (23 [7%] vs 18 [6%]), asthenia (16 [5%] vs 8 [3%]), bone pain (16 [5%] vs 5 [2%]), and febrile neutropenia (16 [5%] vs 9 [3%]). Serious adverse events were reported in 155 (49%) versus 132 (42%). 27 patients died within 30 days of treatment in the cabazitaxel and prednisone plus custirsen group, seven of which were deemed to be treatment related, versus 17 in the cabazitaxel and prednisone group, eight of which were deemed to be treatment related. Of the 21 deaths reported, 15 were reported as complications related to study treatment, either chemotherapy (eight and three, respectively) or study drug (none and four, respectively). INTERPRETATION: We noted no survival benefit in men with metastatic castration-resistant prostate cancer with the addition of custirsen to cabazitaxel and prednisone treatment. Cabazitaxel and prednisone remains the standard of care for patients with metastatic castration-resistant prostate cancer progressing after docetaxel chemotherapy. FUNDING: OncoGenex Pharmaceuticals.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Prednisone/administration & dosage , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/mortality , Taxoids/administration & dosage , Thionucleotides/therapeutic use , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Disease-Free Survival , Docetaxel , Dose-Response Relationship, Drug , Drug Administration Schedule , Humans , Internationality , Kaplan-Meier Estimate , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Invasiveness/pathology , Neoplasm Staging , Prognosis , Proportional Hazards Models , Prospective Studies , Prostatic Neoplasms, Castration-Resistant/diagnostic imaging , Survival Analysis , Thionucleotides/adverse effects , Tomography, X-Ray Computed/methods , Treatment Outcome
6.
Proc Natl Acad Sci U S A ; 110(9): 3495-500, 2013 Feb 26.
Article in English | MEDLINE | ID: mdl-23382200

ABSTRACT

The refractoriness of acute promyelocytic leukemia (APL) with t(11;17)(q23;q21) to all-trans retinoic acid (ATRA)-based therapy concerns clinicians and intrigues basic researchers. By using a murine leukemic model carrying both promyelocytic leukemia zinc finger/retinoic acid receptor-α (PLZF/RARα) and RARα/PLZF fusion genes, we discovered that 8-chlorophenylthio adenosine-3', 5'-cyclic monophosphate (8-CPT-cAMP) enhances cellular differentiation and improves gene trans-activation by ATRA in leukemic blasts. Mechanistically, in combination with ATRA, 8-CPT-cAMP activates PKA, causing phosphorylation of PLZF/RARα at Ser765 and resulting in increased dissociation of the silencing mediator for retinoic acid and thyroid hormone receptors/nuclear receptor corepressor from PLZF/RARα. This process results in changes of local chromatin and transcriptional reactivation of the retinoic acid pathway in leukemic cells. Meanwhile, 8-CPT-cAMP also potentiated ATRA-induced degradation of PLZF/RARα through its Ser765 phosphorylation. In vivo treatment of the t(11;17) APL mouse model demonstrated that 8-CPT-cAMP could significantly improve the therapeutic effect of ATRA by targeting a leukemia-initiating cell activity. This combined therapy, which induces enhanced differentiation and oncoprotein degradation, may benefit t(11;17) APL patients.


Subject(s)
Cell Differentiation , Cyclic AMP/analogs & derivatives , Leukemia, Promyelocytic, Acute/drug therapy , Oncogene Proteins, Fusion/metabolism , Proteolysis , Thionucleotides/therapeutic use , Translocation, Genetic , Tretinoin/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Differentiation/drug effects , Cell Differentiation/genetics , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 17/genetics , Cyclic AMP/pharmacology , Cyclic AMP/therapeutic use , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Humans , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/pathology , Mice , Mice, Inbred C57BL , Nuclear Receptor Co-Repressor 2/metabolism , Phosphorylation/drug effects , Phosphoserine/metabolism , Proteolysis/drug effects , Signal Transduction/drug effects , Survival Analysis , Thionucleotides/pharmacology , Translocation, Genetic/drug effects , Tretinoin/pharmacology
7.
Postepy Hig Med Dosw (Online) ; 70(0): 1182-1189, 2016 Dec 08.
Article in English | MEDLINE | ID: mdl-28026821

ABSTRACT

Survivin is a member of the family of apoptosis inhibitors. It regulates several essential cellular processes, i.e. it inhibits apoptosis and promotes cell proliferation, DNA repair and autophagy. Survivin is responsible for development of the cell's resistance to chemotherapy and radiotherapy. Overexpression of survivin generally correlates with poor prognosis. Its presence has been detected in most types of human tumours. Currently much attention is paid to the possibilities of using this protein as a diagnostic marker of cancer or a prognostic factor. Survivin occurs selectively in cancer cells and is essential for their survival. These features make survivin a promising target for cancer therapy. There are some strategies for discovering survivin inhibitors. The most common strategies are antisense nucleotides, RNA interference and small molecule inhibitors of protein. Scientists are also working on using survivin to induce an immune response in cancer patients. This article discusses the potential role of survivin in the diagnosis of various types of cancer, as well as selected strategies for the inhibition of both gene expression and protein function. Detailed knowledge of the mechanisms of survivin action may therefore be crucial for effective antitumor therapy development.


Subject(s)
Genital Neoplasms, Female/metabolism , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Female , Genital Neoplasms, Female/diagnosis , Genital Neoplasms, Female/drug therapy , Genital Neoplasms, Female/therapy , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Inhibitor of Apoptosis Proteins/genetics , Naphthoquinones/pharmacology , Naphthoquinones/therapeutic use , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/therapeutic use , Prognosis , RNAi Therapeutics , Survivin , Thionucleotides/pharmacology , Thionucleotides/therapeutic use
8.
Blood ; 121(1): 136-47, 2013 Jan 03.
Article in English | MEDLINE | ID: mdl-23165478

ABSTRACT

Several RNA-targeted therapeutics, including antisense oligonucleotides (ONs), small interfering RNAs, and miRNAs, constitute immunostimulatory CpG motifs as an integral part of their design. The limited success with free antisense ONs in hematologic malignancies in recent clinical trials has been attributed to the CpG motif-mediated, TLR-induced prosurvival effects and inefficient target modulation in desired cells. In an attempt to diminish their off-target prosurvival and proinflammatory effects and specific delivery, as a proof of principle, in the present study, we developed an Ab-targeted liposomal delivery strategy using a clinically relevant CD20 Ab (rituximab)-conjugated lipopolyplex nanoparticle (RIT-INP)- and Bcl-2-targeted antisense G3139 as archetypical antisense therapeutics. The adverse immunostimulatory responses were abrogated by selective B cell-targeted delivery and early endosomal compartmentalization of G3139-encapsulated RIT-INPs, resulting in reduced NF-κB activation, robust Bcl-2 down-regulation, and enhanced sensitivity to fludarabine-induced cytotoxicity. Furthermore, significant in vivo therapeutic efficacy was noted after RIT-INP-G3139 administration in a disseminated xenograft leukemia model. The results of the present study demonstrate that CD20-targeted delivery overcomes the immunostimulatory properties of CpG-containing ON therapeutics and improves efficient gene silencing and in vivo therapeutic efficacy for B-cell malignancies. The broader implications of similar approaches in overcoming immunostimulatory properties of RNA-directed therapeutics in hematologic malignancies are also discussed.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antimetabolites, Antineoplastic/therapeutic use , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Molecular Targeted Therapy , Nanoparticles/therapeutic use , Oligonucleotides, Antisense/therapeutic use , Thionucleotides/therapeutic use , Vidarabine/analogs & derivatives , Adjuvants, Immunologic/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antimetabolites, Antineoplastic/pharmacokinetics , Cell Line, Tumor/transplantation , CpG Islands , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Leukemic/drug effects , Gene Silencing , Genes, bcl-2/drug effects , Humans , Liposomes , Mice , Mice, Inbred NOD , Mice, SCID , Nanoparticles/administration & dosage , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Oligonucleotides, Antisense/pharmacokinetics , Proto-Oncogene Proteins c-bcl-2/biosynthesis , RNA, Small Interfering/pharmacology , Rituximab , Thionucleotides/pharmacokinetics , Toll-Like Receptor 9/antagonists & inhibitors , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology , Vidarabine/pharmacokinetics , Vidarabine/therapeutic use , Xenograft Model Antitumor Assays
9.
Semin Cutan Med Surg ; 33(2): 76-82, 2014 Jun.
Article in English | MEDLINE | ID: mdl-25085666

ABSTRACT

Merkel cell carcinoma is an aggressive neuroendocrine tumor with a high incidence of local recurrence, regional nodal and distant metastasis, and a high mortality rate. It has been linked to a polyomavirus in addition to immune suppression. Traditionally, treatment options have been limited to surgery and radiation therapy. Better understanding of the molecular pathways of infection and carcinogenesis has provided potential molecular targets and potential immunotherapies which are discussed in this review.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Merkel Cell/therapy , Immunotherapy , Skin Neoplasms/therapy , Antibodies, Monoclonal/therapeutic use , Benzamides/therapeutic use , Biomarkers/analysis , CD56 Antigen/immunology , Carcinoma, Merkel Cell/diagnosis , Carcinoma, Merkel Cell/epidemiology , Carcinoma, Merkel Cell/virology , Electrochemotherapy , Hepatitis A Virus Cellular Receptor 2 , Humans , Imatinib Mesylate , Immunotherapy, Adoptive , Indazoles , Inhibitor of Apoptosis Proteins/metabolism , Interferons/therapeutic use , Interleukin-12/genetics , Interleukin-2/therapeutic use , Ipilimumab , Lymphatic Metastasis , Membrane Proteins/antagonists & inhibitors , Merkel cell polyomavirus , Oligonucleotides, Antisense , Piperazines/therapeutic use , Polyomavirus Infections/virology , Prognosis , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Pyrimidines/therapeutic use , Receptors, Somatomedin/antagonists & inhibitors , Skin Neoplasms/diagnosis , Skin Neoplasms/epidemiology , Skin Neoplasms/virology , Somatostatin/analogs & derivatives , Sulfonamides/therapeutic use , Survivin , TOR Serine-Threonine Kinases/antagonists & inhibitors , Thionucleotides/therapeutic use
10.
Exp Mol Pathol ; 94(1): 137-47, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23064047

ABSTRACT

With the advent of advanced tools in molecular biology, understanding on cancer etiology has improved. siRNA can be considered as an effective tool in cancer therapy through silencing overexpressed genes responsible for cell proliferation or preventing apoptosis. However, some contentious issues such as stability and delivery of siRNA are to be resolved. Bcl-2, an anti-apoptotic gene, is overexpressed in a wide variety of cancers and responsible for drug resistance tumors. In our earlier studies, we developed a nanoformulation of siRNA targeting the Bcl-2 and achieved successful delivery in vitro and in vivo. To extend the scope of the study further, in the present work, we studied the role of nanoformulation of siRNA as adjuvant in chemotherapy with cisplatin. Dose dependant nephrotoxicity is a serious concern apart from other adverse effects of cisplatin. The IC(50) value for cisplatin was decreased from 9.83 µmol/l to 7.43 µmol/l in HeLa cells and from 8.54 µmol/l to 6.68 µmol/l in HEp-2 cells, when it was given with siRNA nanoformulation. Cisplatin at the dose of 1.7 mg/kg in combination with siRNA nanoformulation was effective in improving the lifespan of tumor bearing mice with significant decrease in nephrotoxicity.


Subject(s)
Antineoplastic Agents/therapeutic use , Cisplatin/therapeutic use , Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Small Interfering/therapeutic use , Thionucleotides/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Chemotherapy, Adjuvant , Cisplatin/pharmacology , Combined Modality Therapy , HeLa Cells , Hep G2 Cells , Humans , Mice , RNA, Small Interfering/pharmacology , Thionucleotides/pharmacology
11.
Curr Oncol Rep ; 15(2): 113-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23266703

ABSTRACT

Adenocarcinoma of the prostate is the most common cancer in men in the Western Hemisphere. This diagnosis includes a clinicopathologically diverse collection of disease entities, encompassing a spectrum from early localized disease to advanced-stage castration-sensitive and ultimately metastatic, castration-resistant states. Although early-stage disease is treatable and potentially curable, treatment options for castration-resistant prostate cancer, the common pathway to prostate cancer death, remain limited and palliative in nature. Therapeutic resistance to androgen blockade, cytotoxic chemotherapy, and radiotherapy is underpinned by a number of cellular mechanisms. The upregulation of protective, antiapoptotic chaperone proteins is one of these mechanisms, and is exemplified by the protein clusterin in castration-resistant prostate cancer. Antisense oligonucleotide technology provides the potential to inhibit specific genes in cancer cells and with this the possibility of a vast impact in oncology, but no antisense drugs have been approved for use in cancer patients to date. Custirsen (OGX-011) is a novel antisense oligonucleotide drug which targets clusterin expression, and its application in prostate cancer is reviewed in this article.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Clusterin/antagonists & inhibitors , Prostatic Neoplasms/drug therapy , Thionucleotides/therapeutic use , Adenocarcinoma/secondary , Clinical Trials as Topic , Humans , Male , Prostatic Neoplasms/secondary
12.
Cancer Invest ; 30(5): 415-21, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22571342

ABSTRACT

BACKGROUND: In carcinogenesis, methylation of DNA promoter regions results in inactivation of tumor-suppressing genes. MG98 was designed to inhibit DNA methyltransferases enzyme 1 production. METHODS: This multicenter study explored two schedules of MG98 with Interferon-α-2ß to identify schedule and dose for patients with metastatic RCC. RESULTS: Doses of IFN 9 MIU/MG98 125 mg/m(2) for a continuous schedule and IFN 9 MIU/MG98 200 mg/m(2) for an intermittent schedule were considered the MTDs. Treatment resulted in one PR and eight SD. CONCLUSION: MG98 combined with IFN was safe and resulted in clinical activity.


Subject(s)
Carcinoma, Renal Cell/drug therapy , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , Kidney Neoplasms/drug therapy , Oligodeoxyribonucleotides/therapeutic use , Thionucleotides/therapeutic use , Adult , Aged , Carcinoma, Renal Cell/mortality , DNA (Cytosine-5-)-Methyltransferase 1 , Disease-Free Survival , Humans , Kidney Neoplasms/mortality , Middle Aged , Oligodeoxyribonucleotides/adverse effects , Thionucleotides/adverse effects
13.
BJU Int ; 110(5): 658-67, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22429837

ABSTRACT

UNLABELLED: What's known on the subject? and What does the study add? Treatment options in the UK for men with metastatic castration-resistant prostate cancer (mCRPC) have been limited, and there is no standard approach, particularly in the second-line setting. The absence of a standard approach is further confounded by the differing definitions and terminologies still used in clinical practice to describe this group of patients (e.g. androgen-independent prostate cancer, hormone refractory prostate cancer, CRPC). With multiple new treatment options emerging, it will be critical to identify key considerations in our decision-making process and to establish an optimum, standardized approach to treatment so that new therapies can be assimilated into an mCRPC treatment algorithm and our routine clinical practice. Most UK oncologists consider patients with advanced, symptomatic prostate cancer as eligible for chemotherapy, although a poor performance status, significant co-morbid factors, advancing age, and the presence of asymptomatic disease with slowly rising prostate-specific antigen levels would prevent chemotherapy use. The decision to retreat with chemotherapy is largely driven by prior response to first-line chemotherapy. Many UK oncologists feel that UK clinical practice is likely to change over the next 5 years, with abiraterone acetate, MDV3100 and cabazitaxel likely to have the most positive impacts in the treatment of mCRPC. OBJECTIVES: To evaluate the current management of patients with advanced prostate cancer by UK oncologists. To gain insights into the future role of emerging therapies. MATERIALS AND METHODS: A semi-structured questionnaire was issued by the British Uro-oncology Group to society members during a closed meeting in September 2010. Emerging therapies evaluated were: abiraterone acetate, aflibercept, bevacizumab, cabazitaxel, custirsen, MDV3100, sipuleucel-T and zibotentan. RESULTS: Eighty of 98 (82%) surveys were completed. Responders had on average 189 new referrals, and treated 126 patients with advanced prostate cancer each year. Chemotherapy was used by 86% of responders for patients with symptomatic metastatic castration-resistant prostate cancer (mCRPC), although poor performance status, advancing age and slowly rising prostate-specific antigen levels would prevent chemotherapy use. The decision to retreat with chemotherapy was largely driven by prior response to first-line chemotherapy, with docetaxel preferred for those responding. Many (78%) felt that UK clinical practice was likely to change over the next 5 years, and that abiraterone acetate, MDV3100 and cabazitaxel would have the most positive impact. Opinions regarding the future use of aflibercept and custirsen were mixed. Few (≤3%) would use zibotentan or bevacizumab in the future based on recent negative phase III study results, or because of cost and complexity for sipuleucel-T. CONCLUSIONS: Although emerging therapies for mCRPC mean that the future is bright, guidelines are needed to ensure optimum use and sequencing of treatments. Additional costs and anticipated workload associated with new agents will require careful consideration.


Subject(s)
Androgen Antagonists/therapeutic use , Antineoplastic Agents/therapeutic use , Practice Patterns, Physicians'/trends , Prostatic Neoplasms/drug therapy , Urology/trends , Androstenes , Androstenols/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Attitude of Health Personnel , Benzamides , Bevacizumab , Forecasting , Humans , Male , Nitriles , Orchiectomy , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/therapeutic use , Pyrrolidines/therapeutic use , Receptors, Vascular Endothelial Growth Factor , Recombinant Fusion Proteins/therapeutic use , Retreatment , Surveys and Questionnaires , Taxoids/therapeutic use , Therapies, Investigational/trends , Thionucleotides/therapeutic use , Tissue Extracts/therapeutic use
14.
JOP ; 13(4): 358-60, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22797389

ABSTRACT

Despite the enormous advances in clinical research in oncology, the prognosis of pancreatic carcinoma remains poor. The therapeutic options in this type of cancer are very limited, with modest results at present. In the 2012 American Society of Clinical Oncology (ASCO) Annual Meeting, four interesting trials on the second line treatment of pancreatic cancer were presented. The first study (Abstract #4017) with a phase II design suggested that maintenance therapy with sunitinib, after a complete course of standard first line treatment, was feasible and effective while the second phase I/II study (Abstract #4034) evaluated the role of trabedersen, an agent that inhibits TGF-ß2 expression. Finally, the efficacy and toxicity of lapatinib combined with either FOLFOX (Abstract #e14533) or capecitabine (Abstract #e14569) were examined in the second line setting of pancreatic cancer.


Subject(s)
Pancreatic Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Capecitabine , Clinical Trials as Topic , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Fluorouracil/administration & dosage , Fluorouracil/analogs & derivatives , Humans , Lapatinib , Leucovorin/administration & dosage , Oligodeoxyribonucleotides/therapeutic use , Organoplatinum Compounds/administration & dosage , Pancreatic Neoplasms/pathology , Quinazolines/administration & dosage , Thionucleotides/therapeutic use
15.
Biochem Biophys Res Commun ; 410(3): 682-7, 2011 Jul 08.
Article in English | MEDLINE | ID: mdl-21699885

ABSTRACT

We have previously reported the in vitro anti-proliferative effect of 4-thio-uridylate (s(4)UMP) on OCM-1 uveal melanoma cells. Here, we assessed the efficacy of s(4)UMP on JY cells. Treatment of JY cells with s(4)UMP suppressed their colony forming activity and induced apoptosis; healthy human bone marrow granulocyte-macrophage progenitor cells were 14-fold less sensitive to the nucleotide. In vivo effectiveness of s(4)UMP was determined using xenograft SCID mouse model. s(4)UMP decreased the cell number and colony forming activity of the total cell content of the femur of SCID mice transplanted with JY cells without affecting the bone marrow of healthy mice. These results suggest that s(4)UMP alone or in combination with other clinically approved anti-leukemic remedies should be further explored as a potential novel therapeutic agent.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia, B-Cell/drug therapy , Thionucleotides/therapeutic use , Uridine Monophosphate/analogs & derivatives , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Mice , Mice, SCID , Stem Cells/drug effects , Thionucleotides/chemistry , Thionucleotides/pharmacokinetics , Uridine Monophosphate/chemistry , Uridine Monophosphate/pharmacokinetics , Uridine Monophosphate/therapeutic use , Xenograft Model Antitumor Assays
16.
J Transl Med ; 9: 125, 2011 Jul 28.
Article in English | MEDLINE | ID: mdl-21798045

ABSTRACT

BACKGROUND: Nucleic acids designed to modulate the expression of target proteins remain a promising therapeutic strategy in several diseases, including cancer. However, clinical success is limited by the lack of efficient intracellular delivery. In this study we evaluated whether electroporation could increase the delivery of antisense oligodeoxynucleotides against bcl-2 (G3139) as well as the efficacy of combination chemotherapy in human melanoma xenografts. METHODS: Melanoma-bearing nude mice were treated i.v. with G3139 and/or cisplatin (DDP) followed by the application of trains of electric pulses to tumors. Western blot, immunohistochemistry and real-time PCR were performed to analyze protein and mRNA expression. The effect of electroporation on muscles was determined by histology, while tumor apoptosis and the proliferation index were analyzed by immunohistochemistry. Antisense oligodeoxynucleotides tumor accumulation was measured by FACS and confocal microscopy. RESULTS: The G3139/Electroporation combined therapy produced a significant inhibition of tumor growth (TWI, more than 50%) accompanied by a marked tumor re-growth delay (TRD, about 20 days). The efficacy of this treatment was due to the higher G3139 uptake in tumor cells which led to a marked down-regulation of bcl-2 protein expression. Moreover, the G3139/EP combination treatment resulted in an enhanced apoptotic index and a decreased proliferation rate of tumors. Finally, an increased tumor response was observed after treatment with the triple combination G3139/DDP/EP, showing a TWI of about 75% and TRD of 30 days. CONCLUSIONS: These results demonstrate that electroporation is an effective strategy to improve the delivery of antisense oligodeoxynucleotides within tumor cells in vivo and it may be instrumental in optimizing the response of melanoma to chemotherapy. The high response rate observed in this study suggest to apply this strategy for the treatment of melanoma patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Electroporation/methods , Melanoma/drug therapy , Oligonucleotides, Antisense/therapeutic use , Thionucleotides/therapeutic use , Xenograft Model Antitumor Assays , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/pharmacology , Cisplatin/therapeutic use , Combined Modality Therapy , Down-Regulation/drug effects , Electrodes , Gene Expression Regulation, Neoplastic/drug effects , Humans , Melanoma/genetics , Melanoma/pathology , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thionucleotides/pharmacology , Treatment Outcome
17.
Invest New Drugs ; 29(5): 971-7, 2011 Oct.
Article in English | MEDLINE | ID: mdl-20349264

ABSTRACT

BACKGROUND: Overexpression of Bcl-2 is associated with worse prognosis for a number of cancer types. The present study was designed to determine the maximum tolerated dose (MTD) of oblimersen (antisense Bcl-2) and gemcitabine when administered to patients with refractory malignancies. MATERIALS AND METHODS: Sixteen patients with advanced solid tumors refractory to standard therapies were treated with escalating doses of oblimersen continuous, 120-h intravenous infusion given every 14 days, with a fixed-dose-rate intravenous infusion of gemcitabine administered on day 5 of each cycle. Serial plasma samples were collected to calculate the pharmacokinetics of oblimersen and gemcitabine, and also to measure the effect of oblimersen on Bcl-2 expression. RESULTS: 7 women and 9 men, median age 55 years (range 35-74 years), received a 5-day infusion of oblimersen at dose levels of 5 mg/kg/day (n = 4) or 7 mg/kg/day (n = 12). On the 5th day of the infusion, gemcitabine was given at 10 mg/m(2)/h for a total dose of 1,000 mg/m(2) (n = 7; cohorts I and II), 1,200 mg/m(2) (n = 3; cohort III), or 1,500 mg/m(2) (n = 6; cohort IV). Edema was the dose-limiting toxicity (DLT), necessitating expansion of cohort IV. No subsequent DLTs were noted. Thus, the maximum planned doses were well tolerated, and a formal MTD was not determined. Most hematologic toxicities were grade 1 or 2. There was low-grade fatigue, nausea/vomiting, and myalgias/arthralgias. Oblimersen C(ss) and AUC increased in relation to the dose escalation, but gemcitabine triphosphate levels did not correlate well with dose. There were no objective responses, though 5 patients had stable disease. A >75% reduction in Bcl-2 expression in peripheral blood mononuclear leucocytes was seen more frequently in patients who achieved stable disease than in progressing patients. CONCLUSIONS: The maximal planned dose levels of oblimersen and gemcitabine in combination were well tolerated. Only one DLT (edema) occurred. There was a correlation between Bcl-2 reduction and stable disease. The recommended doses of the drugs for future studies are 7 mg/kg/day of oblimersen on days 1-5, and gemcitabine 1,500 mg/m(2) on day 5, every two weeks.


Subject(s)
Antineoplastic Agents/therapeutic use , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm , Genes, bcl-2/genetics , Neoplasms/drug therapy , Neoplasms/pathology , Thionucleotides/therapeutic use , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Cohort Studies , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Deoxycytidine/pharmacokinetics , Deoxycytidine/therapeutic use , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Neoplasm Staging , Neoplasms/genetics , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/adverse effects , Oligonucleotides, Antisense/pharmacokinetics , Oligonucleotides, Antisense/therapeutic use , Thionucleotides/administration & dosage , Thionucleotides/adverse effects , Thionucleotides/pharmacokinetics , Treatment Outcome , Gemcitabine
18.
Pediatr Int ; 53(4): 524-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21342350

ABSTRACT

BACKGROUND: Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD) are caused by abnormalities in the DMD gene. The majority of DMD patients have out-of-frame deletion(s), which disrupt the reading frame; while some cases of DMD are caused by duplication or nonsense mutation(s). Most patients with BMD have in-frame deletion(s), which preserve the reading frame. The phenotype of BMD is generally milder than that of DMD. Antisense morpholino-mediated exon skipping, which changes out-of-frame deletions to in-frame deletions, is a promising therapeutic approach for DMD. It is necessary, however, to confirm the exon-skipping event in cells of DMD patients before the clinical trial. METHODS: Fibroblasts isolated from four DMD patients were induced to differentiate into the myogenic lineage by infection with Ad.CAGMyoD. The cells were then transfected with two types of morpholino. The exon-skipping event was analyzed on reverse transcription-polymerase chain reaction. RESULTS: Morpholino B30, which is located at the splicing enhancer of exon 51 of the DMD gene, yielded the desired exon 51-skipping event in all deletion patterns of cells tested. Morpholino I25, which is located at the exon donor, induced two different exon-skipping patterns, which are total or partial exon 51-skipping events. According to the sequence analysis, the unexpected unskipped regions were the 95 bp section and the 188 bp section of exon 51, showing that the cryptic splicing donor was newly produced with I25. Unfortunately, these cryptic splicing donors gave rise to out-of-frame patterns. Based on these in vitro results, B30 would presumably be an effective therapy. Interestingly, the cocktail of B30 and I25 appeared to yield a more efficient exon 51-skipping event. CONCLUSION: An in vitro system was developed that could easily screen the effectiveness of antisense sequences and identify good candidates for therapy with morpholino.


Subject(s)
Exons , Genetic Therapy , Morpholinos/therapeutic use , Muscular Dystrophy, Duchenne/therapy , Oligonucleotides, Antisense/therapeutic use , Sequence Deletion , Clinical Trials as Topic , DNA/genetics , Fibroblasts , Humans , In Vitro Techniques , Muscular Dystrophy, Duchenne/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Thionucleotides/therapeutic use , Transfection
19.
Pharm Stat ; 10(3): 213-7, 2011.
Article in English | MEDLINE | ID: mdl-20568099

ABSTRACT

Evaluation (or assessment)-time bias can arise in oncology trials that study progression-free survival (PFS) when randomized groups have different patterns of timing of assessments. Modelling or computer simulation is sometimes used to explore the extent of such bias; valid results require building such simulations under realistic assumptions concerning the timing of assessments. This paper considers a trial that used a logrank test where computer simulations were based on unrealistic assumptions that severely overestimated the extent of potential bias. The paper shows that seemingly small differences in assumptions can lead to dramatic differences in the apparent operating characteristics of logrank tests.


Subject(s)
Bias , Models, Theoretical , Randomized Controlled Trials as Topic , Research Design , Antineoplastic Agents/therapeutic use , Antineoplastic Agents, Alkylating/therapeutic use , Computer Simulation , Dacarbazine/therapeutic use , Disease-Free Survival , Humans , Kaplan-Meier Estimate , Melanoma/drug therapy , Melanoma/epidemiology , Outcome and Process Assessment, Health Care , Reproducibility of Results , Thionucleotides/therapeutic use , Time Factors , Treatment Outcome , United States , United States Food and Drug Administration
20.
Blood Adv ; 5(13): 2775-2787, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34251414

ABSTRACT

Overexpression of B-cell leukemia/lymphoma 2 (BCL2) renders acute myeloid leukemia (AML) cells resistant to chemotherapy and has been associated with unfavorable outcomes. Oblimersen (G3139) is a phosphorothioate 18-mer antisense oligonucleotide directed against the first 6 BCL2 codons. In a phase 1 study of AML patients treated with G3139, cytarabine, and daunorubicin induction with cytarabine consolidation, no antisense-related toxicity was reported, and BCL2 downregulation occurred in patients achieving complete remission. In this phase 3 trial, untreated older AML patients were randomized to cytarabine (100 mg/m2 per day on days 4-10) and daunorubicin (60 mg/m2 per day on days 4-6) followed by cytarabine consolidation (2000 mg/m2 per day on days 4-8) with (arm A) or without (arm B) G3139 (7 mg/m2 per day on days 1-10 [induction] or days 1-8 [consolidation]). A total of 506 patients were enrolled. No differences in toxicity were observed between arms. Estimated overall survival (OS) at 1 year was 43% for arm A and 40% for arm B (1-sided log rank P = .13), with no differences in disease-free (DFS; P = .26) or event-free survival (P = .80). Subgroup analyses showed patients age <70 years in arm A had improved OS by 1 month vs those in arm B (P = .04), and patients with secondary AML in arm A had better DFS vs those in arm B (P = .04). We conclude that addition of G3139 to chemotherapy failed to improve outcomes of older AML patients. However, more effective means of inhibiting BCL2 are showing promising results in combination with chemotherapy in AML. This trial was registered at www.clinicaltrials.gov as #NCT00085124.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Leukemia, Myeloid, Acute , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cytarabine/therapeutic use , Humans , Leukemia, Myeloid, Acute/drug therapy , Thionucleotides/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL