Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Control Release ; 368: 413-429, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38431093

ABSTRACT

Exosomes continue to attract interest as a promising nanocarrier drug delivery technology. They are naturally derived nanoscale extracellular vesicles with innate properties well suited to shuttle proteins, lipids, and nucleic acids between cells. Nonetheless, their clinical utility is currently limited by several major challenges, such as their inability to target tumor cells and a high proportion of clearance by the mononuclear phagocyte system (MPS) of the liver and spleen. To overcome these limitations, we developed "Smart Exosomes" that co-display RGD and CD47p110-130 through CD9 engineering (ExoSmart). The resultant ExoSmart demonstrates enhanced binding capacity to αvß3 on pancreatic ductal adenocarcinoma (PDAC) cells, resulting in amplified cellular uptake in in vitro and in vivo models and increased chemotherapeutic efficacies. Simultaneously, ExoSmart significantly reduced liver and spleen clearance of exosomes by inhibiting macrophage phagocytosis via CD47p110-130 interaction with signal regulatory proteins (SIRPα) on macrophages. These studies demonstrate that an engineered exosome drug delivery system increases PDAC therapeutic efficacy by enhancing active PDAC targeting and prolonging circulation times, and their findings hold tremendous translational potential for cancer therapy while providing a concrete foundation for future work utilizing novel peptide-engineered exosome strategies.


Subject(s)
Carcinoma, Pancreatic Ductal , Exosomes , Pancreatic Neoplasms , Humans , Exosomes/metabolism , CD47 Antigen , Cell Line, Tumor , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/pathology
2.
PLoS One ; 13(5): e0193644, 2018.
Article in English | MEDLINE | ID: mdl-29851957

ABSTRACT

The KRAS oncogene, present in over 90% of pancreatic ductal adenocarcinomas, is most frequently the result of one of three gain-of-function substitution mutations of codon 12 glycine. Thus far, RAS mutations have been clinically refractory to both direct and selective inhibition by systemic therapeutics. This report presents the results of pre-clinical assessment of a lipoplex comprising a plasmid-encoded, modular bi-functional shRNA (bi-shRNA), which executes selective and multi-mutant allelic KRASG12mut gene silencing, encased within a fusogenic liposome systemic delivery vehicle. Using both a dual luciferase reporter system and a Restriction Fragment Length Polymorphism (RFLP) assay, selective discrimination of KRASG12mut from KRASwt was confirmed in vitro in PANC1 cells. Subsequently, systemic administration of the bi-shRNAKRAS fusogenic lipoplex into female athymic Nu/Nu mice bearing PANC1 xenografts demonstrated intratumoral plasmid delivery, KRASG12mut knockdown, and inhibition of tumor growth, without adverse effect. Clinical trials with the bi-shRNA lipoplex have been implemented.


Subject(s)
Carcinoma, Pancreatic Ductal/prevention & control , Drug Delivery Systems , Mutation , Pancreatic Neoplasms/prevention & control , Proto-Oncogene Proteins p21(ras)/genetics , RNA, Small Interfering/genetics , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Proliferation , Female , Humans , Liposomes , Mice , Mice, Nude , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Plasmids/administration & dosage , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
J Pediatr Hematol Oncol ; 39(4): e183-e186, 2017 05.
Article in English | MEDLINE | ID: mdl-28338569

ABSTRACT

Ewing sarcoma is a highly resistant disease with a <10% chance of survival at 5 years after failure of frontline chemotherapy. This is a case report of an Ewing sarcoma patient with metastatic disease recurrence <2 years after standard chemotherapy/radiation who achieved a durable and sustained complete response after 2 series of treatments with Vigil (GMCSF/bi-shRNA furin DNA autologous tumor immunotherapy) serially manufactured from first and second recurrences with ELISPOT assay correlation. Results support justification of further testing of Vigil with ELISPOT assay as a biomarker to assess level of immune response and correlation with disease control.


Subject(s)
Benzhydryl Compounds/administration & dosage , Cancer Vaccines/administration & dosage , Immunotherapy/methods , Sarcoma, Ewing/therapy , Adolescent , Benzhydryl Compounds/therapeutic use , Cancer Vaccines/therapeutic use , Enzyme-Linked Immunospot Assay , Female , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Humans , Modafinil , RNA, Small Interfering/therapeutic use , Salvage Therapy/methods , Sarcoma, Ewing/diagnosis , Treatment Outcome
4.
Toxicol Sci ; 155(2): 400-408, 2017 02.
Article in English | MEDLINE | ID: mdl-27815492

ABSTRACT

Stathmin-1 (STMN1) is a microtubule-destabilizing protein which is overexpressed in cancer. Its overexpression is associated with poor prognosis and also serves as a predictive marker to taxane therapy. We have developed a proprietary bi-functional shRNA (bi-shRNA) platform to execute RNA interference (RNAi)-mediated gene silencing and a liposome-carrier complex to systemically deliver the pbi-shRNA plasmids. In vitro and in vivo testing demonstrated efficacy and specificity of pbi-shRNA plasmid in targeting STMN1 (Phadke, A. P., Jay, C. M., Wang, Z., Chen, S., Liu, S., Haddock, C., Kumar, P., Pappen, B. O., Rao, D. D., Templeton, N. S., et al. (2011). In vivo safety and antitumor efficacy of bifunctional small hairpin RNAs specific for the human Stathmin 1 oncoprotein. DNA Cell Biol. 30, 715-726.). Biodistribution and toxicology studies in bio-relevant Sprague Dawley rats with pbi-shRNA STMN1 lipoplex revealed that the plasmid DNA was delivered to a broad distribution of organs after a single subcutaneous injection. Specifically, plasmid was detected within the first week using QPCR (threshold 50 copies plasmid/1 µg genomic DNA) at the injection site, lung, spleen, blood, skin, ovary (limited), lymph nodes, and liver. It was not detected in the heart, testis or bone marrow. No plasmid was detected from any organ 30 days after injection. Treatment was well tolerated. Minimal inflammation/erythema was observed at the injection site. Circulating cytokine response was also examined by ELISA. The IL-6 levels were induced within 6 h then declined to the vehicle control level 72 h after the injection. TNFα induction was transiently observed 4 days after the DNA lipoplex treatment. In summary, the pbi-shRNA STMN1 lipoplex was well tolerated and displayed broad distribution after a single subcutaneous injection. The pre-clinical data has been filed to FDA and the pbi-shRNA STMN1 lipoplex is being investigated in a phase I clinical study.


Subject(s)
RNA Interference , RNA, Small Interfering/administration & dosage , Stathmin/antagonists & inhibitors , Stathmin/genetics , Animals , Female , Humans , Injections, Subcutaneous , Interleukin-6/blood , Male , Neoplasms/metabolism , Neoplasms/therapy , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacokinetics , Rats , Rats, Sprague-Dawley , Stathmin/administration & dosage , Stathmin/metabolism , Tissue Distribution
5.
Mol Ther ; 24(8): 1412-22, 2016 08.
Article in English | MEDLINE | ID: mdl-27166877

ABSTRACT

The EWS/FLI1 fusion gene is well characterized as a driver of Ewing's sarcoma. Bi-shRNA EWS/FLI1 is a functional plasmid DNA construct that transcribes both siRNA and miRNA-like effectors each of which targets the identical type 1 translocation junction region of the EWS/FLI1 transcribed mRNA sequence. Previous preclinical and clinical studies confirm the safety of this RNA interference platform technology and consistently demonstrate designated mRNA and protein target knockdown at greater than 90% efficiency. We initiated development of pbi-shRNA EWS/FLI1 lipoplex (LPX) for the treatment of type 1 Ewing's sarcoma. Clinical-grade plasmid was manufactured and both sequence and activity verified. Target protein and RNA knockdown of 85-92% was demonstrated in vitro in type 1 human Ewing's sarcoma tumor cell lines with the optimal bi-shRNA EWS/FLI1 plasmid. This functional plasmid was placed in a clinically tested, liposomal (LP) delivery vehicle followed by in vivo verification of activity. Type 1 Ewing's sarcoma xenograft modeling confirmed dose related safety and tumor response to pbi-shRNA EWS/FLI1 LPX. Toxicology studies in mini-pigs with doses comparable to the demonstrated in vivo efficacy dose resulted in transient fever, occasional limited hypertension at low- and high-dose assessment and transient liver enzyme elevation at high dose. These results provide the justification to initiate clinical testing.


Subject(s)
Liposomes , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Protein c-fli-1/genetics , RNA, Small Interfering/genetics , RNA-Binding Protein EWS/genetics , Sarcoma, Ewing/genetics , Animals , Binding Sites , Cell Line, Tumor , Cytokines/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Gene Knockdown Techniques , Gene Order , Genetic Therapy , Humans , Inflammation Mediators , Male , Oncogene Proteins, Fusion/administration & dosage , Oncogene Proteins, Fusion/chemistry , Plasmids/administration & dosage , Plasmids/genetics , Proto-Oncogene Protein c-fli-1/administration & dosage , Proto-Oncogene Protein c-fli-1/chemistry , RNA Interference , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/chemistry , RNA-Binding Protein EWS/administration & dosage , RNA-Binding Protein EWS/chemistry , Sarcoma, Ewing/mortality , Sarcoma, Ewing/pathology , Sarcoma, Ewing/therapy , Targeted Gene Repair , Transfection , Tumor Burden , Xenograft Model Antitumor Assays
6.
Mol Ther ; 24(8): 1478-83, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27109631

ABSTRACT

Ewing's sarcoma is a devastating rare pediatric cancer of the bone. Intense chemotherapy temporarily controls disease in most patients at presentation but has limited effect in patients with progressive or recurrent disease. We previously described preliminary results of a novel immunotherapy, FANG (Vigil) vaccine, in which 12 advanced stage Ewing's patients were safely treated and went on to achieve a predicted immune response (IFNγ ELISPOT). We describe follow-up through year 3 of a prospective, nonrandomized study comparing an expanded group of Vigil-treated advanced disease Ewing's sarcoma patients (n = 16) with a contemporaneous group of Ewing's sarcoma patients (n = 14) not treated with Vigil. Long-term follow-up results show a survival benefit without evidence of significant toxicity (no ≥ grade 3) to Vigil when administered once monthly by intradermal injection (1 × 10e(6) cells/injection to 1 × 10e(7) cells/injection). Specifically, we report a 1-year actual survival of 73% for Vigil-treated patients compared to 23% in those not treated with Vigil. In addition, there was a 17.2-month difference in overall survival (OS; Kaplan-Meier) between the Vigil (median OS 731 days) and no Vigil patient groups (median OS 207 days). In conclusion, these results supply the rational for further testing of Vigil in advanced stage Ewing's sarcoma.

7.
Mol Ther ; 23(6): 1123-1130, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25619726

ABSTRACT

Stathmin1 (STMN1) is a microtubule modulator that is expressed in multiple cancers and correlates with poor survival. We previously demonstrated in vivo safety of bifunctional (bi) shRNA STMN1 bilamellar invaginated vesicle (BIV) and that systemic delivery correlated with antitumor activity. Patients with superficial advanced refractory cancer with no other standard options were entered into trial. Study design involved dose escalation (four patients/cohort) using a modified Fibonacci schema starting at 0.7 mg DNA administered via single intratumoral injection. Biopsy at baseline, 24/48 hours and resection 8 days after injection provided tissue for determination of cleavage product using next-generation sequencing (NGS) and reverse transcription quantitative polymerase chain reaction (RT-qPCR), 5' RLM rapid amplification of cDNA ends (RACE) assay. Serum pharmacokinetics of circulating plasmid was done. Twelve patients were entered into three dose levels (0.7, 1.4, 7.0 mg DNA). No ≥ grade 3 toxic effects to drug were observed. Maximum circulating plasmid was detected at 30 seconds with less than 10% detectable in all subjects at 24 hours. No toxic effects were observed. Predicted cleavage product was detected by both NGS (n = 7/7 patients analyzed, cohorts 1, 2) and RLM RACE (n = 1/1 patients analyzed cohort 3). In conclusion, bi-shRNA STMN1 BIV is well tolerated and detection of mRNA target sequence-specific cleavage product confirmed bi-shRNA BIV mechanism of action.


Subject(s)
Neoplasms/therapy , RNA, Small Interfering/metabolism , Stathmin/genetics , Tubulin Modulators/pharmacology , Aged , Aged, 80 and over , Dose-Response Relationship, Drug , Drug Delivery Systems , Female , High-Throughput Nucleotide Sequencing , Humans , Injections, Intralesional , Male , Middle Aged , Neoplasms/genetics , Plasmids/blood , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Stathmin/pharmacokinetics , Toxicity Tests
8.
Discov Med ; 15(81): 101-10, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23449112

ABSTRACT

Despite billions of dollars allocated to cancer research, cancer remains the number 2 cause of death in the United States with less than 50% of advanced cancer patients living one year following standard treatment. Cancer is a complex disease both intrinsically and in relation to its host environment. From a molecular standpoint no two cancers are the same despite histotypic similarity. As evidenced by the recent advances in molecular biology, treatment for advanced cancer is headed towards specific targeting of vulnerable signaling nodes within the reconfigured pathways created by "omic" rewiring. With advancements in proteo-genomics and the capacity of bioinformatics, complex tumor biology can now be more effectively and rapidly analyzed to discover the vulnerable high information transfer nodes within individual tumors. RNA interference (RNAi) technology, with its capability to knock down the expression of targeted genes (the vulnerable nodes), is moving into the clinic to target these nodes, which are integral to tumor maintenance, with a low risk of side-effects and to block intrinsic immunosuppressors thereby priming the tumor for immune attack. An RNAi based sequential approach, a so called "one-two punch," is being advocated comprising tumor volume reduction (ideally to minimal residual disease status) effected by integrated multi-target knockdown followed by immune activation. Examples and recent developments are provided to illustrate this highly powerful approach heralding the future of personalized cancer therapy.


Subject(s)
Neoplasms/therapy , Precision Medicine , RNA Interference , Clinical Trials as Topic , Humans
9.
Methods Mol Biol ; 942: 259-78, 2013.
Article in English | MEDLINE | ID: mdl-23027056

ABSTRACT

The discovery of RNA interference (RNAi) engendered great excitement and raised expectations regarding its potential applications in biomedical research and clinical usage. Over the ensuing years, expanded understanding of RNAi and preliminary results from early clinical trials tempered enthusiasm with realistic appraisal resulting in cautious optimism and a better understanding of necessary research and clinical directions. As a result, data from more recent trials are beginning to show encouraging positive clinical outcomes. The capability of delivering a pharmacologically effective dose to the target site while avoiding adverse host reactions still remains a challenge although the delivery technology continues to improve. We have developed a novel vector-driven bifunctional short hairpin RNA (bi-shRNA) technology that harnesses both cleavage-dependent and cleavage-independent RISC loading pathways to enhance knockdown potency. Consequent advantages provided by the bi-shRNA include a lower effective systemic dose than comparator siRNA/shRNA to minimize the potential for off-target side effects, due to its ability to induce both a rapid (inhibition of protein translation) and delayed (mRNA cleavage and degradation) targeting effect depending on protein and mRNA kinetics, and a longer duration of effectiveness for clinical applications. Here, we provide an overview of key molecular methods for the design, construction, quality control, and application of bi-shRNA that we believe will be useful for others interested in utilizing this technology.


Subject(s)
Genetic Engineering/methods , RNA, Small Interfering/genetics , Base Sequence , Cholesterol/metabolism , DNA/genetics , DNA/metabolism , Fatty Acids, Monounsaturated/metabolism , Gene Knockdown Techniques , HEK293 Cells , Humans , Inverted Repeat Sequences , Quaternary Ammonium Compounds/metabolism , RNA Cleavage , RNA Interference , Transfection
10.
PLoS One ; 7(8): e40452, 2012.
Article in English | MEDLINE | ID: mdl-22905092

ABSTRACT

Pancreatic and duodenal homeobox-1 (PDX-1) is a transcription factor that regulates insulin expression and islet maintenance in the adult pancreas. Our recent studies demonstrate that PDX-1 is an oncogene for pancreatic cancer and is overexpressed in pancreatic cancer. The purpose of this study was to demonstrate that PDX-1 is a therapeutic target for both hormonal symptoms and tumor volume in mouse models of pancreatic cancer, insulinoma and islet neoplasia. Immunohistochemistry of human pancreatic and islet neoplasia specimens revealed marked PDX-1 overexpression, suggesting PDX-1 as a "drugable" target within these diseases. To do so, a novel RNA interference effector platform, bifunctional shRNA(PDX-1), was developed and studied in mouse and human cell lines as well as in mouse models of pancreatic cancer, insulinoma and islet neoplasia. Systemic delivery of bi-shRNA(humanPDX-1) lipoplexes resulted in marked reduction of tumor volume and improved survival in a human pancreatic cancer xenograft mouse model. bi-shRNA(mousePDX-1) lipoplexes prevented death from hyperinsulinemia and hypoglycemia in an insulinoma mouse model. shRNA(mousePDX-1) lipoplexes reversed hyperinsulinemia and hypoglycemia in an immune-competent mouse model of islet neoplasia. PDX-1 was overexpressed in pancreatic neuroendocrine tumors and nesidioblastosis. These data demonstrate that PDX-1 RNAi therapy controls hormonal symptoms and tumor volume in mouse models of pancreatic cancer, insulinoma and islet neoplasia, therefore, PDX-1 is a potential therapeutic target for these pancreatic diseases.


Subject(s)
Homeodomain Proteins/metabolism , Insulinoma/therapy , Islets of Langerhans/metabolism , Pancreatic Neoplasms/therapy , RNA Interference , Trans-Activators/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Glucose Tolerance Test , Humans , Immunohistochemistry/methods , In Situ Nick-End Labeling , Insulin/metabolism , Male , Mice , Mice, SCID , Neoplasm Transplantation , Pancreas/metabolism
11.
Mol Ther ; 20(3): 679-86, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22186789

ABSTRACT

We performed a phase I trial of FANG vaccine, an autologous tumor-based product incorporating a plasmid encoding granulocyte-macrophage colony-stimulating factor (GMCSF) and a novel bifunctional short hairpin RNAi (bi-shRNAi) targeting furin convertase, thereby downregulating endogenous immunosuppressive transforming growth factors (TGF) ß1 and ß2. Patients with advanced cancer received up to 12 monthly intradermal injections of FANG vaccine (1 × 10(7) or 2.5 × 10(7) cells/ml injection). GMCSF, TGFß1, TGFß2, and furin proteins were quantified by enzyme-linked immunosorbent assay (ELISA). Safety and response were monitored. Vaccine manufacturing was successful in 42 of 46 patients of whom 27 received ≥1 vaccine. There were no treatment-related serious adverse events. Most common grade 1, 2 adverse events included local induration (n = 14) and local erythema (n = 11) at injection site. Post-transfection mean product expression GMCSF increased from 7.3 to 1,108 pg/10(6) cells/ml. Mean TGFß1 and ß2 effective target knockdown was 93.5 and 92.5% from baseline, respectively. Positive enzyme-linked immunospot (ELISPOT) response at month 4 was demonstrated in 9 of 18 patients serially assessed and correlated with survival duration from time of treatment (P = 0.025). Neither dose-adverse event nor dose-response relationship was noted. In conclusion, FANG vaccine was safe and elicited an immune response correlating with prolonged survival. Phase II assessment is justified.


Subject(s)
Cancer Vaccines/therapeutic use , Furin/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Neoplasms/therapy , RNA, Small Interfering/therapeutic use , Adult , Aged , Cancer Vaccines/administration & dosage , Cancer Vaccines/adverse effects , Female , Gene Expression , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Interferon-gamma/biosynthesis , Leukocytes, Mononuclear/immunology , Male , Middle Aged , Neoplasm Staging , Neoplasms/immunology , Neoplasms/mortality , Neoplasms/pathology , Survival Analysis , Transgenes , Treatment Outcome
12.
Pharm Res ; 28(12): 2983-95, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22009588

ABSTRACT

Since its discovery in 1998, RNA interference (RNAi) has revolutionized basic and clinical research. Small RNAs, including small interfering RNA (siRNA), short hairpin RNA (shRNA) and microRNA (miRNA), mediate RNAi effects through either cleavage-dependent or cleavage-independent RNA inducible silencing complex (RISC) effector processes. As a result of its efficacy and potential, RNAi has been elevated to the status of "blockbuster therapeutic" alongside recombinant protein and monoclonal antibody. RNAi has already contributed to our understanding of neoplasia and has great promise for anti-cancer therapeutics, particularly so for personalized cancer therapy. Despite this potential, several hurdles have to be overcome for successful development of RNAi-based pharmaceuticals. This review will discuss the potential for, challenges to, and the current status of RNAi-based cancer therapeutics.


Subject(s)
MicroRNAs/therapeutic use , Neoplasms/genetics , Neoplasms/therapy , RNA Interference , RNA, Small Interfering/therapeutic use , Animals , Genetic Therapy , Humans , MicroRNAs/administration & dosage , MicroRNAs/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics
13.
DNA Cell Biol ; 30(9): 715-26, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21612405

ABSTRACT

Bifunctional small hairpin RNAs (bi-shRNAs) are functional miRNA/siRNA composites that are optimized for posttranscriptional gene silencing through concurrent mRNA cleavage-dependent and -independent mechanisms (Rao et al., 2010 ). We have generated a novel bi-shRNA using the miR30 scaffold that is highly effective for knockdown of human stathmin (STMN1) mRNA. STMN1 overexpression well documented in human solid cancers correlates with their poor prognosis. Transfection with the bi-shSTMN1-encoding expression plasmid (pbi-shSTMN1) markedly reduced CCL-247 human colorectal cancer and SK-Mel-28 melanoma cell growth in vitro (Rao et al., 2010 ). We now examine in vivo the antitumor efficacy of this RNA interference-based approach with human tumor xenografted athymic mice. A single intratumoral (IT) injection of pbi-shSTMN1 (8 µg) reduced CCL-247 tumor xenograft growth by 44% at 7 days when delivered as a 1,2-dioleoyl-3-trimethyl-ammoniopropane:cholesterol liposomal complex. Extended growth reductions (57% at day 15; p < 0.05) were achieved with three daily treatments of the same construct. STMN1 protein reduction was confirmed by immunoblot analysis. IT treatments with pbi-shSTMN1 similarly inhibited the growth of tumorgrafts derived from low-passage primary melanoma (≥70% reduction for 2 weeks) and abrogated osteosarcoma tumorgraft growth, with the mature bi-shRNA effector molecule detectable for up to 16 days after last injection. Antitumor efficacy was evident for up to 25 days posttreatment in the melanoma tumorgraft model. The maximum tolerated dose by IT injection of >92 µg (Human equivalent dose [HED] of >0.3 mg/kg) in CCL-247 tumor xenograft-bearing athymic mice was ∼10-fold higher than the extrapolated IC(50) of 9 µg (HED of 0.03 mg/kg). Healthy, immunocompetent rats were used as biorelevant models for systemic safety assessments. The observed maximum tolerated dose of <100 µg for intravenously injected pbi-shSTMN1 (mouse equivalent of <26.5 µg; HED of <0.09 mg/kg) confirmed systemic safety of the therapeutic dose, hence supporting early-phase assessments of clinical safety and preliminary efficacy.


Subject(s)
Colorectal Neoplasms/therapy , Gene Knockdown Techniques/methods , Genetic Therapy/methods , Melanoma/therapy , RNA Interference , RNA, Small Interfering/metabolism , Stathmin/metabolism , Animals , DNA Primers/genetics , Female , Humans , Immunoblotting , Male , Maximum Tolerated Dose , Mice , Mice, Nude , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
14.
World J Surg ; 35(8): 1700-14, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21557010

ABSTRACT

Normal cellular survival is dependent on the cooperative expression of genes' signaling through a broad array of DNA patterns. Cancer, however, has an Achilles' heel. Its altered cellular survival is dependent on a limited subset of signals through mutated DNA, possibly as few as three. Identification and control of these signals through the use of RNA interference (RNAi) technology may provide a unique clinical opportunity for the management of cancer that employs genomic-proteomic profiling to provide a molecular characterization of the cancer, leading to targeted therapy customized to an individual cancer signal. Such an approach has been described as "personalized therapy." The present review identifies unique developing technology that employs RNAi as a method to target, and therefore block, signaling from mutated DNA and describes a clinical pathway toward its development in cancer therapy.


Subject(s)
Genotype , Neoplasms/genetics , Neoplasms/therapy , Precision Medicine/trends , RNA Interference/physiology , Forecasting , Genetic Predisposition to Disease/genetics , Humans
15.
Expert Rev Mol Med ; 12: e26, 2010 Aug 18.
Article in English | MEDLINE | ID: mdl-20716384

ABSTRACT

RNA interference (RNAi) is an evolutionary conserved mechanism for specific gene silencing. This mechanism has great potential for use in targeted cancer therapy. Understanding the RNAi mechanism has led to the development of several novel RNAi-based therapeutic approaches currently in the early phases of clinical trials. It remains difficult to effectively deliver the nucleic acids required in vivo to initiate RNAi, and intense effort is under way in developing effective and targeted systemic delivery systems for RNAi. Description of in vivo delivery systems is not the focus of this review. In this review, we cover the rationale for pursuing personalised cancer therapy with RNAi, briefly review the mechanism of each major RNAi therapeutic technique, summarise and sample recent results with animal models applying RNAi for cancer, and provide an update on current clinical trials with RNAi-based therapeutic agents for cancer therapy. RNAi-based cancer therapy is still in its infancy, and there are numerous obstacles and issues that need to be resolved before its application in personalised therapy focusing on patient-cancer-specific targets can become standard cancer treatment, either alone or in combination with other treatments.


Subject(s)
Neoplasms/therapy , RNA Interference/physiology , Clinical Trials as Topic , Humans , MicroRNAs/genetics , Models, Biological , Neoplasms/genetics , RNA, Small Interfering/genetics
16.
Gene Regul Syst Bio ; 3: 103-4, 2009 Jun 19.
Article in English | MEDLINE | ID: mdl-19838337

ABSTRACT

Elevated Dicer and Drosha mRNA levels have been documented across a range of tumor types (including ovarian carcinoma) by a number of investigators without any demonstrable correlation with patient survival nor evidence of interference with shRNA processing. A recent publication by Merritt et al. (NEJM 359(25):2641-50, 2008) reporting their findings in patients with ovarian carcinoma reach opposite conclusions. Further study will be needed to resolve this issue.

17.
Adv Drug Deliv Rev ; 61(9): 746-59, 2009 Jul 25.
Article in English | MEDLINE | ID: mdl-19389436

ABSTRACT

RNA interference (RNAi) is a natural process through which expression of a targeted gene can be knocked down with high specificity and selectivity. Using available technology and bioinformatics investigators will soon be able to identify relevant bio molecular tumor network hubs as potential key targets for knockdown approaches. Methods of mediating the RNAi effect involve small interfering RNA (siRNA), short hairpin RNA (shRNA) and bi-functional shRNA. The simplicity of siRNA manufacturing and transient nature of the effect per dose are optimally suited for certain medical disorders (i.e. viral injections). However, using the endogenous processing machinery, optimized shRNA constructs allow for high potency and sustainable effects using low copy numbers resulting in less off-target effects, particularly if embedded in a miRNA scaffold. Bi-functional design may further enhance potency and safety of RNAi-based therapeutics. Remaining challenges include tumor selective delivery vehicles and more complete evaluation of the scope and scale of off-target effects. This review will compare siRNA, shRNA and bi-functional shRNA.


Subject(s)
Genetic Therapy , Neoplasms/therapy , RNA Interference , RNA, Small Interfering/genetics , Animals , Humans
18.
J Neurosci Methods ; 143(2): 95-106, 2005 Apr 30.
Article in English | MEDLINE | ID: mdl-15814141

ABSTRACT

The cellular heterogeneity of brain tissue presents a challenge to gene expression profiling of specific neuronal cell types. The present study employed a fluorescent neural tracer to specifically label midbrain dopamine neurons and non-dopamine cortical neurons. The labeled cells were then used to visually guide harvesting of the cells by laser capture microdissection (LCM). RNA extracted from the two populations of harvested cells was then amplified, labeled and co-hybridized to high density cDNA microarrays for two-color differential expression profiling. Many of the genes most highly enriched in the dopamine neurons were found to be genes previously known to define the dopamine neuronal phenotype. However, results from the microarray were only partially validated by quantitative RT-PCR analysis. The results indicate that LCM harvesting of specific neuronal phenotypes can be effectively guided in a complex cellular environment by specific pre-labeling of the target cell populations and underlie the importance of independent validation of microarray results.


Subject(s)
Cell Separation/methods , Dopamine/metabolism , Gene Expression Profiling/methods , Microdissection/methods , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Oligonucleotide Array Sequence Analysis/methods , Animals , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Lasers , Male , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction/methods , Spectrometry, Fluorescence/methods , Staining and Labeling/methods , Stilbamidines , Systems Integration
19.
Exp Cell Res ; 290(2): 346-57, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14567992

ABSTRACT

Bone marrow stroma fibroblastoid cells (BMSFC) develop from a single clone of cells within each of the in vitro fibroblastoid colonies (CFU-F) derived from either murine or human bone marrow. All of the clones represented by these colonies displayed antigenic and product markers for osteoblast, smooth muscle, and adipocyte lineages when tested separately for each marker. Separate sets of fibroblastoid colonies derived from the same individual donor's culture tested positive with antibodies specific for smooth muscle-specific heavy chain myosin (SMMHC), smooth muscle alpha actin-1, bone sialoprotein, osteocalcin, or alkaline phosphatase, and developed von Kossa-positive deposits shown by X-ray microanalysis and electron diffraction to be hydroxyapatite. Individual cells were positive for both SMMHC and osteocalcin. All cells in the multiple clones tested were capable of metabolizing a fatty acid to form intracellular lipid droplets. PCR transcripts obtained from the human cell cultures that provided these BMSFC clones were consistent with the immunocytochemical findings. Transcripts for PPAR (gamma)-2 and Cbfa-1 were dependent upon the culture medium content, suggesting an osteoblast/adipocyte differentiation switch point. Cell lineage specificity for markers and RNA transcripts was determined by comparison to skin fibroblast controls. These findings demonstrate a high degree of interlineage plasticity in vitro for BMSFC.


Subject(s)
Adipocytes/cytology , Bone Marrow Cells , Cell Lineage , Muscle, Smooth/cytology , Osteoblasts/cytology , Stromal Cells/cytology , Adolescent , Adult , Animals , Biomarkers/analysis , Cells, Cultured , Clone Cells/cytology , Fibroblasts/cytology , Flow Cytometry , Glucose/metabolism , Humans , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Middle Aged , Polymerase Chain Reaction , RNA/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...