Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Oncology ; 87(1): 21-9, 2014.
Article in English | MEDLINE | ID: mdl-24968881

ABSTRACT

Therapies for advanced hepatocellular carcinoma (HCC) are limited. We carried out a phase I trial of a novel autologous whole-cell tumor cell immunotherapy (FANG™), which incorporates a dual granulocyte macrophage colony-stimulating factor (GM-CSF) expressive/bifunctional small hairpin RNA interference (bi-shRNAi) vector. The bi-shRNAi DNA targets furin, which is a proconvertase of transforming growth factors beta (TGFß) 1 and 2. Safety, mechanism, immunoeffectiveness, and suggested benefit were previously shown [Senzer et al.: Mol Ther 2012;20:679-689; Senzer et al.: J Vaccines Vaccin 2013;4:209]. We now provide further follow-up of a subset of 8 HCC patients. FANG manufacturing was successful in 7 of 8 attempts (one failure due to insufficient cell yield). Median GM-CSF expression was 144 pg/10(6) cells, TGFß1 knockdown was 100%, and TGFß2 knockdown was 93% of the vector-transported cells. Five patients were vaccinated (1 or 2.5×10(7) cells/intradermal injection, 6-11 vaccinations). No FANG toxicity was observed. Three of these patients demonstrated evidence of an immune response to the autologous tumor cell sample. Long-term follow-up demonstrated survival of 319, 729, 784, 931+, and 1,043+ days of the FANG-treated patients. In conclusion, evidence supports further assessment of the FANG immunotherapy in HCC.


Subject(s)
Cancer Vaccines/administration & dosage , Carcinoma, Hepatocellular/therapy , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Liver Neoplasms/therapy , RNA, Small Interfering/genetics , Vaccination , Adult , Aged , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Female , Gene Expression , Gene Knockdown Techniques , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Humans , Injections, Intradermal , Liver Neoplasms/immunology , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , Middle Aged , Survival Analysis , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta2/genetics , Transforming Growth Factor beta2/metabolism , Transplantation, Autologous , Treatment Outcome
2.
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
3.
Mol Ther ; 18(2): 429-34, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19935775

ABSTRACT

A phase I clinical trial was conducted to determine the clinical safety of Telomelysin, a human telomerase reverse transcriptase (hTERT) promoter driven modified oncolytic adenovirus, in patients with advanced solid tumors. A single intratumoral injection (IT) of Telomelysin was administered to three cohorts of patients (1 x 10(10), 1 x 10(11), 1 x 10(12) viral particles). Safety, response and pharmacodynamics were evaluated. Sixteen patients with a variety of solid tumors were enrolled. IT of Telomelysin was well tolerated at all dose levels. Common grade 1 and 2 toxicities included injection site reactions (pain, induration) and systemic reactions (fever, chills). hTERT expression was demonstrated at biopsy in 9 of 12 patients. Viral DNA was transiently detected in plasma in 13 of 16 patients. Viral DNA was detectable in four patients in plasma or sputum at day 7 and 14 post-treatment despite below detectable levels at 24 h, suggesting viral replication. One patient had a partial response of the injected malignant lesion. Seven patients fulfilled Response Evaluation Criteria in Solid Tumors (RECIST) definition for stable disease at day 56 after treatment. Telomelysin was well tolerated. Evidence of antitumor activity was suggested.


Subject(s)
Adenoviridae/metabolism , Neoplasms/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/metabolism , Telomerase/metabolism , Adenoviridae/genetics , Adult , Aged , Female , Flow Cytometry , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasms/genetics , Neoplasms/metabolism , Oncolytic Virotherapy/adverse effects , Oncolytic Viruses/genetics , Telomerase/genetics
4.
J Gene Med ; 12(5): 403-12, 2010 May.
Article in English | MEDLINE | ID: mdl-20440751

ABSTRACT

BACKGROUND: Hereditary inclusion body myopathy (HIBM) is an autosomal recessive adult onset myopathy. It is characterized by mutations of the GNE (UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase) gene. Afflicted patients have no therapeutic options. In preclinical testing, we have previously demonstrated the ability to correct GNE gene function and the safety of delivery of wild type GNE gene using a liposomal delivery vehicle. METHODS: A single patient (subject #001) with severe HIBM treated by compassionate investigational new drug received four doses of GNE gene Lipoplex via intramuscular injection. GNE transgene expression, downstream induction of sialic acid, safety and muscle function were evaluated. RESULTS: Significant durable improvement in locoregional skeletal muscle function was observed in the injected left extensor carpi radialis longus of #001 in correlation with GNE transgene upregulation and local induction of sialic acid. Other than transient low grade fever and pain at the injection site, no significant toxicity was observed. CONCLUSIONS: Proof of principle for manufacturing of 'clinical grade' GNE gene Lipoplex, clinical safety and activity are demonstrated with GNE gene Lipoplex. Further assessment will involve intravenous administration and subsequent phase I trial involving additional but less severely afflicted HIBM patients.


Subject(s)
Genetic Therapy , Liposomes/metabolism , Multienzyme Complexes/genetics , Multienzyme Complexes/therapeutic use , Myositis, Inclusion Body/genetics , Myositis, Inclusion Body/therapy , Adolescent , Adult , Biopsy , Female , Genetic Therapy/adverse effects , Humans , Injections, Intramuscular , Muscle Strength , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Myositis, Inclusion Body/physiopathology , N-Acetylneuraminic Acid/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use , Young Adult
5.
Curr Gene Ther ; 9(1): 45-60, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19275571

ABSTRACT

Tremendous strides have been made in proteogenomics and RNA interference technologies. Hence "personalized" cancer gene therapy has become a foreseeable rather than a predictable reality. Currently, the lack of an optimized, systemic gene delivery vehicle remains a key limiting factor for developing effective treatment applications. Since their introduction by Felgner in 1987, cationic lipids have been an attractive consideration for gene delivery, in view of their biocompatibility, biodegradability, low toxicity, and low immunogenicity. Successful in vivo transgene expression by cationic lipid- or cationic polymer-based delivery depends critically on a long circulating half life (>48 h), a definable systemic biodistribution with target-specific cancer localization, and efficient cell entry and internalization. Ideally, the agent should have a hydrophobic, stabilized core that ensures integrity of the therapeutic entity in vivo, a biocompatible, neutrally charged shell (zeta potential of approximately +/-10 mv) for enhanced, "stealth" circulation, and a suitable size (approximately 50-200 nm in diameter) for access into the tumor neovasculature and reduced reticuloendothelial system (RES) uptake. "Smart" receptor-targeting moieties can redirect intracellular trafficking. Additional engineered features have also been incorporated to minimize lysosomal degradation (membrane fusogenic lipids or proton sponge), promote endosomal escape into cytoplasm (cell penetrating peptides, triblock copolymer construction), and enhance nuclear entry and activate the endogenous transcriptional machinery (inclusion of a nuclear localization signal). Improvements in each of these respective areas of study have converged to yield promising in vivo results.


Subject(s)
Gene Transfer Techniques , Genetic Therapy , Genetic Vectors , Lipids/administration & dosage , Neoplasms/therapy , Animals , Humans , Neoplasms/genetics
6.
ISRN Oncol ; 2012: 247657, 2012.
Article in English | MEDLINE | ID: mdl-22523703

ABSTRACT

Ewing's sarcoma tumors are associated with chromosomal translocation between the EWS gene and the ETS transcription factor gene. These unique target sequences provide opportunity for RNA interference(i)-based therapy. A summary of RNAi mechanism and therapeutically designed products including siRNA, shRNA and bi-shRNA are described. Comparison is made between each of these approaches. Systemic RNAi-based therapy, however, requires protected delivery to the Ewing's sarcoma tumor site for activity. Delivery systems which have been most effective in preclinical and clinical testing are reviewed, followed by preclinical assessment of various silencing strategies with demonstration of effectiveness to EWS/FLI-1 target sequences. It is concluded that RNAi-based therapeutics may have testable and achievable activity in management of Ewing's sarcoma.

7.
Clin Cancer Res ; 17(1): 183-92, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21208907

ABSTRACT

PURPOSE: On the basis of the hypothesis that the combined expression of immunostimulatory granulocyte macrophage colony stimulating factor (GM-CSF) and antitumor suppressor TGF-ß2 antisense (AS) transgenes can break tolerance and stimulate immune responses to cancer-associated antigens, we constructed an expression plasmid [the tumor-associated glycoprotein (TAG) plasmid] that coexpresses GM-CSF and TGF-ß2 AS nucleotide sequences and which was incorporated into an autologous whole-cell vaccine. EXPERIMENTAL DESIGN: Patients undergoing resection were enrolled. Freshly harvested autologous tumor cells were mechanically and enzymatically disaggregated, then electroporated with the TAG vector. The resulting vaccine was irradiated, then aliquoted and cryopreserved until the time of injection. Patients received a minimum of 5 to a maximum of 12 monthly intradermal injections. Immune function was monitored at baseline and at months 3 and 6. RESULTS: Vaccine manufacturing efficiency was 84% (32/38). Twenty-three patients received at least 1 vaccination. There were no grade 3 or 4 toxicities, and grade 1 and 2 events were local in nature. Seventeen of 21 patients had stable disease (SD) at month 2 or later as their best response, and 1 patient with stage IVa malignant melanoma achieved a complete response (CR) following 11 vaccinations and remains without evidence of disease 2 years following initiation of therapy. Six of 13 patients displayed a positive enzyme-linked immunospot (ELISPOT) response to autologous TAG vaccine at week 12 including 3 patients with prolonged SD or CR. The 3 other patients survived through week 24, as compared with none of the 7 ELISPOT-negative patients. CONCLUSIONS: On the basis of safety and clinical and immunologic results, further evaluation of bifunctional vaccines is warranted.


Subject(s)
Cancer Vaccines/genetics , Cancer Vaccines/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Neoplasms/therapy , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/therapeutic use , Transforming Growth Factor beta2/genetics , Adult , Aged , Aged, 80 and over , Cancer Vaccines/administration & dosage , Cancer Vaccines/adverse effects , Female , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Male , Middle Aged , Neoplasm Staging , Neoplasms/immunology , Transplantation, Autologous
8.
Hum Gene Ther ; 22(11): 1331-41, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21517694

ABSTRACT

Hereditary inclusion body myopathy (HIBM) is an autosomal recessive adult-onset myopathy due to mutations in the GNE (UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase) gene. Affected patients have no therapeutic options. We have previously demonstrated in preclinical testing the ability to safely correct GNE gene function through liposomal delivery of the wild-type GNE gene. Results were verified in a single patient treated by intravenous infusion of GNE gene lipoplex. A single patient (patient 001) with severe HIBM treated with a compassionate investigational new drug received seven doses of GNE gene lipoplex via intravenous infusion at the following doses: 0.4, 0.4, 1.0, 4.0, 5.0, 6.0, and 7.0 mg of DNA. GNE transgene expression, downstream induction of sialic acid, safety, and muscle function were evaluated. Transient low-grade fever, myalgia, tachycardia, transaminase elevation, hyponatremia, and hypotension were observed after infusion of each dose of GNE gene lipoplex. Quadriceps muscle expression of the delivered GNE, plasmid, and RNA was observed 24 hr after the 5.0-mg dose and at significantly greater levels 72 hr after the 7.0-mg infusion in comparison with expression in quadriceps muscle immediately before infusion. Sialic acid-related proteins were increased and stabilization in the decline of muscle strength was observed. We conclude that clinical safety and activity have been demonstrated with intravenous infusion of GNE gene lipoplex. Further assessment will involve a phase I trial of intravenous administration of GNE gene lipoplex in individuals with less advanced HIBM with more muscle function.


Subject(s)
Multienzyme Complexes/genetics , Myositis, Inclusion Body/therapy , Adult , Female , Genetic Therapy , Genetic Vectors , Humans , Infusions, Intravenous , Liposomes , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Myositis, Inclusion Body/genetics , RNA/metabolism
9.
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
10.
Gene Regul Syst Bio ; 3: 181-90, 2009 Oct 21.
Article in English | MEDLINE | ID: mdl-20054407

ABSTRACT

Hereditary inclusion body myopathy type 2 (HIBM2) is a myopathy characterized by progressive muscle weakness with early adult onset. The disease is the result of a recessive mutation in the Glucosamine (UDP-N-acetyl)-2-epimerase/N-acetylmannosamine kinase gene (GNE), which results in reduced enzyme function and sialic acid levels. A majority of individuals with HIBM2 are from Iranian-Jewish or Japanese decent, but isolated cases have been identified world wide. This article reviews the diagnostic criteria for HIBM2. Current research with a highlight on the biology of the disease and the role of GNE in the sialic acid pathway are assessed. Finally, therapeutic investigations and animal models are discussed with a focus on future studies to better understand the pathology of Hereditary Inclusion Body Myopathy and move therapeutic agents towards clinical trials.

11.
Gene Regul Syst Bio ; 3: 89-101, 2009 May 08.
Article in English | MEDLINE | ID: mdl-19838336

ABSTRACT

Hereditary inclusion body myopathy-2 (HIBM2) is an adult-onset, muscular disease caused by mutations in the GNE gene. HIBM2-associated GNE mutations causing hyposialyation have been proposed to contribute to reduced muscle function in patients with HIBM2, though the exact cause of this disease is unknown. In the current studies we examined pre-clinical in vivo toxicity, and expression of the plasmid-based, CMV driven wild-type GNE plasmid vector. The plasmid vector was injected intramuscularly (IM) or systemically (IV) into BALB/c mice, following encapsulation in a cationic liposome (DOTAP:Cholesterol). Single IM injections of the GNE-lipoplex at 40 microg did not produce overt toxicity or deaths, indicating that the no observable adverse effect level (NOAEL) dose for IM injection was >or=40 microg. Single intravenous (IV) infusion of GNE-lipoplex was lethal in 33% of animals at 100 microg dose, with a small proportion of animals in the 40 microg cohort demonstrating transient toxicity. Thus the NOAEL dose by the IV route was greater than 10 microg and less than or equal to 40 microg. Real-time RT-qPCR analysis demonstrated recombinant human GNE mRNA expression in 100% of muscle tissues that received IM injection of 40 microg GNE-lipoplex, at 2 weeks. These results indicate that GNE-lipoplex gene transfer is safe and can produce durable transgene expression in treated muscles. Our findings support future exploration of the clinical efficacy of GNE-lipoplex for experimental gene therapy of HIBM2.

12.
Expert Rev Anticancer Ther ; 8(9): 1461-70, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18759697

ABSTRACT

Stathmin 1 (STMN1), also known as p17, p18, p19, 19K, metablastin, oncoprotein 18, LAP 18 and Op18, is a 19 kDa cytosolic protein. It was the first discovered member of a family of phylogenetically related microtubule-destabilizing phosphoproteins critically involved in the construction and function of the mitotic spindle. A threshold level of STMN1 is required for orderly progression through mitosis in a variety of cell types. STMN1 is overexpressed across a broad range of human malignancies (leukemia, lymphoma, neuroblastoma; ovarian, prostatic, breast and lung cancers and mesothelioma). It is also upregulated in normally proliferating cell lines but is only rarely upregulated in nonproliferating cell lines with the exception of neurons, anterior pituitary cells and glial cells. Its expression is also upregulated in hepatocytes during regeneration and in lymphoid cells when they are signaled to proliferate. In this review, we summarize available data as rationale for the therapeutic manipulation of STMN1 in cancer patients.


Subject(s)
Neoplasms/metabolism , Stathmin/biosynthesis , Genetic Therapy , Humans , Mitosis , Neoplasms/therapy , Spindle Apparatus/physiology , Stathmin/genetics , Up-Regulation
13.
Gene Regul Syst Bio ; 2: 243-52, 2008 Jun 20.
Article in English | MEDLINE | ID: mdl-19787087

ABSTRACT

Hereditary Inclusion Body Myopathy (HIBM2) is a chronic progressive skeletal muscle wasting disorder which generally leads to complete disability before the age of 50 years. There is currently no effective therapeutic treatment for HIBM2. Development of this disease is related to expression in family members of an autosomal recessive mutation of the GNE gene, which encodes the bifunctional enzyme UDP-GlcNAc 2-epimerase/ManNAc kinase (GNE/MNK). This is the rate limiting bifunctional enzyme that catalyzes the first 2 steps of sialic acid biosynthesis. Decreased sialic acid production, consequently leads to decreased sialyation of a variety of glycoproteins including the critical muscle protein alpha-dystroglycan (alpha-DG). This in turn severely cripples muscle function and leads to the onset of the syndrome. We hypothesize that replacing the mutated GNE gene with the wildtype gene may restore functional capacity of GNE/MNK and therefore production of sialic acid, allowing for improvement in muscle function and/or delay in rate of muscle deterioration. We have constructed three GNE gene/CMV promoter plasmids (encoding the wildtype, HIBM2, and Sialuria forms of GNE) and demonstrated enhanced GNE gene activity following delivery to GNE-deficient CHO-Lec3 cells. GNE/MNK enzyme function was significantly increased and subsequent induction of sialic acid production was demonstrated after transfection into Lec3 cells with the wild type or R266Q mutant GNE vector. These data form the foundation for future preclinical and clinical studies for GNE gene transfer to treat HIBM2 patients.

14.
J Clin Oncol ; 26(27): 4418-25, 2008 Sep 20.
Article in English | MEDLINE | ID: mdl-18802154

ABSTRACT

PURPOSE: Generation of broad cytotoxic T-lymphocyte responses against multiple epitopes and tumor-associated antigens (TAAs) may provide effective immunotherapy in patients with cancer. We evaluated a single-vial peptide vaccine consisting of nine HLA-A2 supertype-binding epitopes (two native and seven analog epitopes modified for optimal HLA binding or T-cell receptor stimulation) covering five TAAs and the universal helper pan-DR epitope, formulated as a stable emulsion with incomplete Freund's adjuvant (Montanide ISA 51; Seppic SA, Paris, France). The clinical efficacy, safety, and multiepitope immunogenicity of IDM-2101 was evaluated in patients with stage IIIB or IV non-small-cell lung cancer (NSCLC). PATIENTS AND METHODS: A total of 63 patients were enrolled who were positive for HLA-A2. End points included survival, safety, and immune response. IDM-2101 (previously EP-2101) was administered every 3 weeks for the first 15 weeks, then every 2 months through year 1, then quarterly through year 2, for a total of 13 doses. Epitope-specific cytotoxic and helper T-lymphocyte immunogenic responses were measured by the interferon gamma enzyme-linked immunosorbent spot assay. RESULTS: No significant adverse events were noted. Low-grade erythema and pain at the injection site were the most common adverse effects. One-year survival in the treated patients was 60%, and median survival was 17.3 months. One complete and one partial response were identified. Survival was longer in patients demonstrating an immune response to epitope peptides (P < .001). CONCLUSION: IDM-2101 was well tolerated, and evidence of efficacy was suggested.


Subject(s)
Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Carcinoma, Non-Small-Cell Lung/prevention & control , Carcinoma, Non-Small-Cell Lung/secondary , Lung Neoplasms/immunology , Lung Neoplasms/prevention & control , T-Lymphocytes, Cytotoxic/immunology , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/immunology , Epitopes, T-Lymphocyte/immunology , Female , Humans , Male , Middle Aged , Remission Induction
15.
J Clin Oncol ; 24(29): 4721-30, 2006 Oct 10.
Article in English | MEDLINE | ID: mdl-16966690

ABSTRACT

PURPOSE: Belagenpumatucel-L is a nonviral gene-based allogeneic tumor cell vaccine that demonstrates enhancement of tumor antigen recognition as a result of transforming growth factor beta-2 inhibition. PATIENTS AND METHODS: We performed a randomized, dose-variable, phase II trial involving stages II, IIIA, IIIB, and IV non-small-cell lung cancer patients. Each patient received one of three doses (1.25, 2.5, or 5.0 x 10(7) cells/injection) of belagenpumatucel-L on a monthly or every other month schedule to a maximum of 16 injections. Immune function, safety, and anticancer activity were monitored. RESULTS: Seventy-five patients (two stage II, 12 stage IIIA, 15 stage IIIB, and 46 stage IV patients) received a total of 550 vaccinations. No significant adverse events were observed. A dose-related survival difference was demonstrated in patients who received > or = 2.5 x 10(7) cells/injection (P = .0069). Focusing on the 61 late-stage (IIIB and IV) assessable patients, a 15% partial response rate was achieved. The estimated probabilities of surviving 1 and 2 years were 68% and 52%, respectively for the higher dose groups combined and 39% and 20%, respectively, for the low-dose group. Immune function was explored in the 61 advanced-stage (IIIB and IV) patients. Increased cytokine production (at week 12 compared with patients with progressive disease) was observed among clinical responders (interferon gamma, P = .006; interleukin [IL] -6, P = .004; IL-4, P = .007), who also displayed an elevated antibody-mediated response to vaccine HLAs (P = .014). Furthermore, positive enzyme-linked immunospot reactions to belagenpumatucel-L showed a correlation trend (P = .086) with clinical responsiveness in patients achieving stable disease or better. CONCLUSION: Belagenpumatucel-L is well tolerated, and the survival advantage justifies further phase III evaluation.


Subject(s)
Cancer Vaccines/therapeutic use , Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/therapy , Oligonucleotides, Antisense/therapeutic use , Transforming Growth Factor beta/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Antibody Formation/drug effects , Cancer Vaccines/adverse effects , Cancer Vaccines/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Female , Humans , Interferon-gamma/metabolism , Lung Neoplasms/immunology , Male , Middle Aged , Oligonucleotides, Antisense/adverse effects , Oligonucleotides, Antisense/immunology , Survival Analysis , Transforming Growth Factor beta2 , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL