Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Cancer Gene Ther ; 16(3): 206-16, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18949015

ABSTRACT

We examined the microRNA (miRNA) expression profile of 40 prostatectomy specimens from stage T2a/b, early relapse and non-relapse cancer patients, to better understand the relationship between miRNA dysregulation and prostate oncogenesis. Paired analysis was carried out with microdissected, malignant and non-involved areas of each specimen, using high-throughput liquid-phase hybridization (mirMASA) reactions and 114 miRNA probes. Five miRNAs (miR-23b, -100, -145, -221 and -222) were significantly downregulated in malignant tissues, according to significance analysis of microarrays and paired t-test with Bonferroni correction. Lowered expression of miR-23b, -145, -221 and -222 in malignant tissues was validated by quantitative reverse transcription (qRT)-PCR analyses. Ectopic expression of these miRNAs significantly reduced LNCaP cancer cell growth, suggesting growth modulatory roles for these miRNAs. Patient subset analysis showed that those with post-surgery elevation of prostate-specific antigen (chemical relapse) displayed a distinct expression profile of 16 miRNAs, as compared with patients with non-relapse disease. A trend of increased expression (>40%) of miR-135b and miR-194 was observed by qRT-PCR confirmatory analysis of 11 patients from each clinical subset. These findings indicate that an altered miRNA expression signature accompanied the prostate oncogenic process. Additional, aberrant miRNA expression features may reflect a tendency for early disease relapse. Growth inhibition through the reconstitution of miRNAs is potentially applicable for experimental therapy of prostate cancer, pending molecular validation of targeted genes.


Subject(s)
Adenocarcinoma/genetics , Gene Expression Profiling , MicroRNAs/analysis , Prostatic Neoplasms/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/surgery , Cell Line, Tumor/chemistry , Disease Progression , Humans , Male , MicroRNAs/biosynthesis , Neoplasm Metastasis , Neoplasm Staging/methods , Postoperative Period , Prostatectomy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/surgery , Reverse Transcriptase Polymerase Chain Reaction
2.
Cancer Gene Ther ; 15(6): 341-55, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18369380

ABSTRACT

MicroRNAs (miRNAs) were discovered more than a decade ago as noncoding, single-stranded small RNAs (approximately 22 nucleotides) that control the timed gene expression pattern in Caenorhabditis elegans life cycle. A number of these evolutionarily conserved, endogenous miRNAs have been shown to regulate mammalian cell growth, differentiation and apoptosis. miRNAs are multispecific by nature. The individual miRNA is capable of modulating the expression of a network of mRNAs that it binds by imperfect sequence complementarity. Human cancers commonly exhibit an altered expression profile of miRNAs with oncogenic (miR-21, miR-106a and miR-155) or tumor-suppressive (let-7, miR-15a/16, miR-34a and miR-143/145) activity. As consistent with the natural function of miRNAs in specifying cellular phenotype, miRNA-based cancer gene therapy offers the theoretical appeal of targeting multiple gene networks that are controlled by a single, aberrantly expressed miRNA. Reconstitution of tumor-suppressive miRNA, or sequence-specific knockdown of oncogenic miRNAs by 'antagomirs,' has produced favorable antitumor outcomes in experimental models. We discuss pending issues that need to be resolved prior to the consideration of miRNA-based experimental cancer gene therapy. These include the need for definitive mRNA target validation, our incomplete understanding of rate-limiting cellular components that impact the efficiency of this posttranscriptional gene-silencing phenomenon, the possibility for nonspecific immune activation and the lack of a defined, optimal mode of delivery.


Subject(s)
Genetic Therapy/methods , MicroRNAs/genetics , Neoplasms/therapy , Humans , MicroRNAs/metabolism , Neoplasms/genetics
3.
Cancer Gene Ther ; 14(11): 885-93, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17704755

ABSTRACT

ONYX-015 is an attenuated chimeric human group C adenovirus, which preferentially replicates in and lyses tumor cells that are p53 negative. The purpose of this phase I, dose-escalation study was to determine the safety and feasibility of intravenous infusion with ONYX-015 in combination with enbrel in patients with advanced carcinoma. Enbrel is a recombinant dimer of human tumor-necrosis factor (TNF)-alpha receptor, previously shown to reduce the level of functional TNF. Nine patients, three in each cohort received multiple cycles of ONYX-015 infusion (1 x 10(10), 1 x 10(11) and 1 x 10(12) vp weekly for 4 weeks/cycle) in addition to subcutaneous enbrel (only during cycle 1) injections per FDA-indicated dosing. Of the nine patients, four had stable disease. No significant adverse events were attributed to the experimental regimen, confirming that enbrel can be safely administered along with oncolytic virotherapy. Two of the three patients in cohort 3 had detectable viral DNA at days 3 and 8 post-ONYX-015 infusion. Their detectable circulating viral DNA was markedly higher during cycle 1 (with enbrel coadministration) as compared with cycle 2 (without enbrel) at the same time points. Area under the curve determinations indicate a marked higher level of TNF-alpha induction and accelerated clearance at cycle 2 in the absence of enbrel. Further assessment is recommended.


Subject(s)
Adenoviridae , Antineoplastic Agents/adverse effects , Carcinoma/drug therapy , Immunoglobulin G/adverse effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Etanercept , Female , Humans , Immunoglobulin G/administration & dosage , Immunoglobulin G/therapeutic use , Infusions, Intravenous , Male , Middle Aged , Receptors, Tumor Necrosis Factor/administration & dosage , Receptors, Tumor Necrosis Factor/therapeutic use , Tumor Necrosis Factor-alpha/blood , Viral Vaccines
4.
Clin Cancer Res ; 7(3): 691-703, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11297266

ABSTRACT

CD40 binding produces multifaceted growth signals in normal and malignant B cells, whereas its physiological role is less well characterized in epithelial cancers. We examined the growth outcome of CD40 ligation in human breast cancer cells, using CD40+ (T47D and BT-20) and CD40-negative (MCF-7, ZR-75-1) cell lines as defined by flow cytometric analysis, immunohistochemistry, and reverse transcription-PCR. Treatment with the soluble recombinant CD40 ligand (CD40L) molecules gp39 or CD40L-trimer significantly reduced [3H]thymidine uptake in BT-20 and T47D cells by up to 40%, but did not affect the growth of CD40-negative MCF-7 or ZR-75-1 cells. Similarly, significant growth inhibition was observed after co-incubation with CD40L-transfected murine L cells (55.0 +/- 8.9%, P < 0.001) that express membrane CD40L constitutively, or with paraformaldehyde-fixed, CD3+ CD40L+ PBLs from three different HLA-mismatched donors (39.7 +/- 3.7%, P < 0.01). Untransfected L cells and non-CD40L-expressing lymphocytes did not produce significant growth inhibition. The in vivo antitumorigenic effects of CD40L were examined using a s.c. severe combined immunodeficient-hu xenograft model. Pretreatment with two different soluble recombinant CD40L constructs (CD40L and gp39) produced similar xenograft growth-inhibitory effects [67 +/- 24% (n = 4), and 65 +/- 14% (n = 8) inhibition, respectively], which were reversed by co-treatment with the CD40L-neutralizing antibody LL48. In vitro analysis indicated that CD40L-induced growth inhibition was accompanied by apoptotic events including cell shrinkage, rounding, and detachment from the adherent T47D culture monolayer. Thirty-one and 27% of gp39-treated T47D and BT-20 cells underwent apoptosis, respectively, as compared with 56 and 65% from the same cell lines after treatment with the Fas agonistic antibody CH-11. An up-regulation of the proapoptotic protein Bax in T47D and BT-20 cells was observed, which indicated that this Bcl-2 family member may contribute to this growth-inhibitory effect. To explore the clinical relevance of CD40L-CD40 interaction, retrospective immunohistochemical analysis was carried to characterize in situ CD40- and CD40L-expression in breast cancer patient biopsies. All of the infiltrating ductal (5 of 5 cases tested) and lobular (4 of 4 cases) breast carcinomas, carcinomas in situ (6 of 6 cases), and mucinous carcinoma tested (1 case) expressed CD40. Varying proportions of tumor cells also expressed CD40L in the majority of infiltrating ductal (3 of 5 cases) and lobular (3 of 4 cases) carcinomas, and carcinomas in situ (4 of 6 cases), as determined by immunohistochemistry and validated by RT-PCR detection of the CD40L message in only CD40L positive-staining cases. Tumor infiltrating mononuclear cells from infiltrating carcinomas and carcinomas in situ expressed CD40 (10 of 10 cases), but less commonly CD40L (1 case of infiltrating lobular carcinoma, 2 cases of carcinoma in situ). Our findings indicate that the CD40 signaling pathway is active in human breast carcinoma cells. However, tumor-infiltrating lymphocytes from primary tumor tissues may be limited in their capacity to directly modulate tumor growth through the CD40L-CD40 loop.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , CD40 Ligand/biosynthesis , CD40 Ligand/pharmacology , Animals , Annexin A5/metabolism , Apoptosis , Blotting, Western , CD40 Antigens/metabolism , Carcinoma/metabolism , Cell Division/drug effects , Dimerization , Flow Cytometry , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Leukocytes, Mononuclear/metabolism , Mice , Mice, SCID , Neoplasm Transplantation , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleases/metabolism , Thymidine/metabolism , Time Factors , Transfection , Tumor Cells, Cultured , Up-Regulation
5.
Clin Lung Cancer ; 2(3): 220-6, 2001 Feb.
Article in English | MEDLINE | ID: mdl-14700482

ABSTRACT

Non-small-cell lung cancer frequently contains oncogenetic defects (mutations in ras, retinoblastoma, and p53 genes) that contribute to disease pathophysiology. Recent studies and clinical trials have focused on gene therapy approaches that either replace the function of defective tumor-suppressor genes such as p53 or inactivate mutant oncogenes such as ras. Ribozymes are RNA molecules with highly specific intrinsic enzymatic activity against target RNA sequences, which can discriminate mutant sequences that differ by a single base from their wild-type counterparts. Following binding to the RNA substrate by base-pair complementation, the ribozyme cleaves the target RNA irreversibly, then releases itself for new rounds of subsequent cleavage, resulting in significantly improved target:effector stoichiometry as compared with antisense oligonucleotides of the same specificity. Transcript-specific ribozymes have been used extensively for experimental oncogene inactivation. Ribozymes are effective for targeting mutant ras, p53, or the multidrug-resistant gene product for lung cancer cells in vitro. However, their in vivo effect is not well defined against this malignancy. We recently characterized the antitumor properties of an anti-K-ras ribozyme specific for the K-ras codon 12 mutation (GGT-->GTT). When delivered as a transgene by an adenoviral vector (ADV), the K-ras ribozyme (KRbz) suppressed growth of lung tumor xenografts expressing the relevant mutation, whereas the corresponding antisense sequence lacking catalytic activity did not. Multiple intratumoral (3-5) injections of KRbz-ADV were effective in producing complete tumor regressions of preexisting tumor xenografts. Clinical trials are under consideration to examine the applicability of this anti-K-ras ribozyme for treatment of non-small-cell lung cancers expressing the relevant mutation.

6.
Am J Clin Oncol ; 23(5): 463-72, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11039505

ABSTRACT

The purpose of this study is to determine immune recovery and function after treatment with docetaxel or paclitaxel. Peripheral blood mononuclear cells were harvested before chemotherapy and at weekly times afterwards for cycle 1. Leukocyte subsets ICD45hiCD14lo polymorphonuclear neutrophils, CD45hiCD14hi monocytes, CD45hiCD14- lymphocytes, CD3+CD4/CD8+ T cells, CD3-CD19+ B cells, CD3-CD16/CD56+ natural killer (NK) cells], and circulating cytokine levels [tumor necrosis factor-alpha, gamma-interferon (gamma-IFN), and interleukins (IL-2, IL-10, IL-12)] were followed. In addition, T-cell mitogenic function, NK function, and lymphokine activated killer (LAK) function was assessed. Ten patients were entered in the trial. T-cell frequency, B-cell frequency, and CD4/CD8 ratio did not change. IL-10 serum levels significantly decreased in paclitaxel-treated patients (4.4+/-1.3 pg/ml at week 4 versus 7.8+/-2.1 pg/ml at baseline; p < 0.05). IL-2, IL-12, and gamma-IFN levels were not detectable. NK cytotoxic activity decreased in docetaxel-treated patients. LAK cell activity was not altered. Four patients achieved a partial or complete response. They demonstrated higher than normal CD4:CD8 T-cell ratios and an improved phytohemagglutinin stimulation index (SI = 2.5). In conclusion, our findings suggest that immune function was affected more significantly after docetaxel treatment. Investigational approaches, which enhance cellular immunity, may be of greater relevance after treatment with docetaxel. Additional studies monitoring NK function after chemotherapy are recommended.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cytokines/blood , Cytotoxicity, Immunologic/drug effects , Immunity, Cellular/drug effects , Killer Cells, Natural , Paclitaxel/analogs & derivatives , Paclitaxel/pharmacology , Taxoids , Aged , Antineoplastic Agents, Phytogenic/therapeutic use , Docetaxel , Female , Humans , Immunophenotyping , Killer Cells, Lymphokine-Activated , Killer Cells, Natural/drug effects , Lymphocyte Activation , Lymphocyte Subsets , Male , Middle Aged , Neoplasms/drug therapy , Neoplasms/immunology , Paclitaxel/therapeutic use
7.
Mol Biotechnol ; 15(1): 39-49, 2000 May.
Article in English | MEDLINE | ID: mdl-10911621

ABSTRACT

ras mutations represent one of the most common oncogenetic lesions in human non-small cell lung cancer (NSCLC) and adversely affect the survival of patients afflicted with this disease. ras-directed gene therapy in the past employed primarily antisense oligonucleotides (AS-ODN) or expression vectors (such as a viral vector construct) that deliver the antisense sequence to inactivate the mutant oncogene message. These approaches produced minimal toxicity, and yet were limited in efficacy. Ribozymes present a viable alternative in antisense therapy by virtue of their renewable catalytic capability for site-specific RNA cleavage. We recently produced an adenoviral vector with a hammerhead ribozyme transgene (KRbz) that is specific for the K-ras codon 12 mutant sequence GUU, given the considerations that (a) in the United States, approx 30% of human NSCLCs express K-ras oncogene mutations, nearly all of which reside in codon 12; (b) anti-K-ras, anti-H, as well as anti-N-ras hammerhead ribozymes are potent growth inhibitors in various human cancers tested; and (c) in vitro and animal model studies suggest that ribozymes directed at oncogene (K- and H-ras C-fos, BCR-ABL) or human immunodeficiency viral gene messages are more effective than their antisense counterpart. This article describes the techniques involved in the production of the KRbz-adenoviral vector that is specific for the K-ras mutation GTT, and summarizes its in vivo antitumor effect against NSCLC xenografts expressing the relevant K-ras mutation in athymic mice.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Genes, ras , Genetic Techniques , Genetic Vectors/genetics , Lung Neoplasms/genetics , RNA, Catalytic/genetics , Cell Line , DNA Mutational Analysis/methods , Fatty Acids, Monounsaturated/chemistry , Humans , Liposomes , Mutation , Plasmids , Quaternary Ammonium Compounds/chemistry , Recombination, Genetic , Sequence Analysis, DNA , Transfection/methods
8.
Leuk Lymphoma ; 36(5-6): 543-58, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10784400

ABSTRACT

We and others previously demonstrated that human multiple myeloma (MM) cells express CD40 and have an active CD40-growth regulatory pathway. This study characterizes the growth outcome of soluble (gp39) or membrane-bound recombinant human CD40-ligand (rCD40L) and its relationship with Fas-dependent apoptosis. Contrary to the moderate growth-stimulatory effect of the CD40-MAb G28.5, gp39 inhibited 3H-thymidine uptake of the plasma dyscrasia lines ARH-77, U266, and HS-Sultan in a dose-dependent fashion by up to 82%. By comparison, RPMI 8226 cells were resistant to CD40L-growth modulation, which may be attributable to a single base substitution (TCA-->TTA, serine-->leucine) at the 3rd cysteine-rich extramembrane region of CD40. Gp39 similarly reduced myeloma clonogenic colony (MCC) formation in patient primary bone marrow cultures by 50% (40-76%; n=6). Studies using transfectant L cells that constitutively expressed CD40L showed that membrane-bound CD40L inhibited the growth of ARH-77, U266, and HS-Sultan cells (66%, 63%, and 32%, respectively), whereas untransfected L cells did not. Growth inhibition by gp39 or CD40L+ L cells was neutralized by coincubation with the CD40L antibodies 5c8 or LL48. CD40L-treatment increased apoptotic activity of MM cells, as defined by oligonucleosomal DNA fragmentation and an increased binding to annexin V (16-28%). All three untreated CD40-responsive MM lines expressed the Fas/Apo-1/CD95 antigen (65-92% CD95+). However, only ARH-77 cells responded to the growth inhibitory effect of the CD95-agonistic antibody CH-11. CD95 expression was not affected significantly by gp39 treatment, and growth inhibition by CH-11 was additive to gp39 (from 42% to 64% decrease in 3H-thmidine uptake). Conversely, the CD95 antagonist antibody ZB4 reversed the Fas-dependent growth inhibitory process but did not significantly alter gp39-mediated growth outcome. Gp39 treatment lowered the expression of TNFR-associated factors TRAF4 and TRAF6 by 38% and 32%, respectively, whereas detectable levels of TRAF1,2,3, and 5 levels remained unchanged. Our observations indicate that the CD40L-binding inhibits human MM cell growth and increases its apoptotic activity. This growth inhibitory effect corresponds to lower levels of cytoplasmic TRAF signaling elements, and appears independent of the Fas-signaling pathway. CD40 receptor mutation may lead to unresponsiveness to CD40 growth modulation in multiple myeloma cells.


Subject(s)
Apoptosis/drug effects , Membrane Glycoproteins/pharmacology , Multiple Myeloma/pathology , fas Receptor/metabolism , CD40 Ligand , Humans , Immunotherapy , Membrane Glycoproteins/metabolism , Multiple Myeloma/immunology , Multiple Myeloma/metabolism , Multiple Myeloma/therapy , Recombinant Proteins/metabolism , Signal Transduction , Tumor Cells, Cultured
9.
Gene Ther ; 7(23): 2041-50, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11175317

ABSTRACT

Approximately 15-30% of human non-small cell lung cancers (NSCLC) carry K-ras mutations, among which point mutations at codon 12 are the most common. This study characterizes the anti-tumor effect of an anti-K-ras ribozyme adenoviral vector (KRbz-ADV; replication-deficient, E1-deleted Ad5 backbone) against NSCLC lines that express the relevant mutation (K-ras codon 12 GGT --> GTT; H441 and H1725). KRbz-ADV significantly inhibited tumor cell growth (38-94% reduction by 3H-thymidine uptake) in a time- and dose-dependent manner, but produced minimal growth inhibition on normal epithelial cells, or NSCLC H1650 cells that lack the relevant mutation. The in vivo anti-tumorigenic effect of KRbz-ADV treatment was characterized with cell line xenografts in nu/nu mice. Pre-treatment with KRbz-ADV (10 or 20 p.f.u. per cell) completely abrogated subcutaneous engraftment of H441 (n = 13) or H1725 cells (n = 8), as compared with a 100% tumor take and progressive tumor growth in animals that received untreated tumor cells, or control vector (luciferase-adenovirus/Luc-ADV)-treated tumor cells. Pre-treatment with a mutant anti-K-ras ribozyme adenoviral vector (mutKRbz-ADV), which has the same specificity as KRbz but lacks ribozyme catalytic activity, did not produce an anti-tumorigenic effect. The in vivo effect of KRbz-ADV treatment was further examined by initiating injections (2 x 10(9) p.f.u.) at 7 days after tumor induction. Pre-existing tumor growth was reduced by 39% by a single intratumoral injection. Repeat injections (three or five KRbz-ADV-intratumoral injections at 2 x 10(9) p.f.u. every other day) resulted in complete tumor regression in five of seven mice. In contrast, single or multiple injections of control vector Luc-ADV did not significantly alter tumor xenograft outcome. Ribozyme expression was confirmed in H441 cells that demonstrated reduced growth after KRbz-ADV treatment. Reduced growth corresponded to significantly lowered levels of K-ras mRNA, as defined by RT-PCR (51% of untreated level, n = 3) and RNase protection assay (56% of untreated level, n = 4) analyses. Further, 37.5% of KRbz-ADV-treated cells underwent apoptosis, as compared with 11.7%, and 19.0% in untreated and Luc-ADV-treated cultures, respectively. A significantly higher proportion of KRbz-ADV-treated H441 cells (58.2%) underwent apoptosis when maintained under anchor-independent conditions that simulate in vivo tumorigenesis ('anoikis'). This is the first report that demonstrates that KRbz-ADV can effectively inhibit in vivo tumorigenesis, and produces regression of pre-existing human lung tumor xenografts having the relevant K-ras mutation.


Subject(s)
Carcinoma, Non-Small-Cell Lung/therapy , Genes, ras , Genetic Therapy/methods , Lung Neoplasms/therapy , RNA, Catalytic/administration & dosage , Adenoviridae/genetics , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Genetic Vectors/administration & dosage , Humans , Lung Neoplasms/genetics , Mice , Mice, Nude , Neoplasm Transplantation , Point Mutation , Transplantation, Heterologous , Tumor Cells, Cultured
10.
Methods Mol Med ; 35: 261-71, 2000.
Article in English | MEDLINE | ID: mdl-21390810

ABSTRACT

Lung cancer is the leading cause of cancer death for men and women in the United States. Several factors affect survival in nonsmall-cell lung cancer [NSCLC; i.e., stage, age, Karnofsky Performance Status (KPS)]. The 5-yr survival rate is<15% for newly diagnosed cases following conventional treatments. Tumor oncogenetic defects appear to be associated with adverse survival (1,2). The presence of a ras mutation or p21(ras) oncoprotein overexpression correlates with an unfavorable prognosis for patients with nonsmall-cell lung cancer (3). These findings suggest that abnormal ras function contributes to pathophysiology, and raise the possibility that reversal of aberrant ras activity may serve as a viable therapeutic approach.

11.
Cell Prolif ; 32(4): 231-8, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10614712

ABSTRACT

As at present only a long-term follow-up can fully determine whether monoclonal gammapathies of undetermined significance (MGUS) will evolve into multiple myeloma (MM), this study attempted to identify other variables connected with the amount of monoclonal component (MC), generally considered as the most reliable marker of malignant evolution. Thirty-four MGUS subjects showing a high MC (> or = 15.0 g/l) but without clinical evidence of MM (MGUS group b), were characterized for their phenotypic and genotypic profile by comparing them either with 40 MM patients or with 24 subjects affected by a benign form of monoclonal gammapathy (MGUS group a) according to the standard criteria. In addition to the usual laboratory markers, the levels of expression of a panel of CD membrane subsets were measured on B and T lymphocytes. Also, the serum level of the p53 mutant protein and the structural alterations of the c-myc oncogene were evaluated. The results show that for MGUS group b patients, an increased M-protein was accompanied by significantly increased levels of peripheral blood CD3+ T cells and oncogenetic aberrations in c-myc. Since a high serum MC level seems to indicate a greater likelihood of malignant transformation for MGUS patients, these findings suggest that this relationship may be a result of the concomitant alterations observed at a phenotypic and genotypic level. Such alterations may be potentially useful as surrogate markers for the transition of benign to malignant (MM) plasma cell dyscrasia.


Subject(s)
Genetic Markers , Monoclonal Gammopathy of Undetermined Significance/genetics , Multiple Myeloma/genetics , Paraproteins/genetics , Adult , Aged , Aged, 80 and over , Biomarkers , Blotting, Southern , DNA, Neoplasm/analysis , Disease Progression , Female , Genes, myc/physiology , Genotype , Humans , Immunophenotyping , Male , Middle Aged , Monoclonal Gammopathy of Undetermined Significance/diagnosis , Multiple Myeloma/diagnosis , Phenotype , Predictive Value of Tests , Prognosis , Transformation, Genetic
12.
Leuk Lymphoma ; 21(1-2): 1-8, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8907262

ABSTRACT

CD40 is a 48 kDa glycosylated phospoprotein that is a member of the tumor necrosis factor receptor (TNF-R) superfamily. CD40 was originally identified in B lymphocytes, and is found on monocytes, dendritic cells, some carcinoma cell lines, and the thymic epithelium. CD40 is expressed on normal pre-B through mature B stages of differentiation. For normal B cells, the cross-linking of CD40 induces cell cycle progression, long-term proliferation in vitro, IgE secretion, increased adhesion molecule (LFA-1) expression, and low level IL-6 secretion. The natural ligand of CD40 (CD40L, gp39, or T-BAM, for T-B cell activating molecule) was recently identified as an inducible molecule expressed transitionally on activated T cells. Although originally believed to be absent in normal and malignant plasma cells, CD40 has been demonstrated on the majority of myeloma cell lines and myeloma cells from plasma cell dyscrasia (PCD) patient specimens tested. CD40 activation modulated myeloma cell proliferation and clonogenicity in vitro, suggesting that the CD40 pathway is active in myeloma cell growth. For the IL-6 dependent cell line ANBL-6, CD40 activation was associated with autocrine IL-6 production. However, the IL-6 pathway does not appear to play a predominant role in CD40 activation of non-IL-6-dependent MM cell lines and patient primary bone marrow cultures. The possible pathophysiologic role of the CD40 receptor in human multiple myeloma is discussed.


Subject(s)
Antibodies/metabolism , CD40 Antigens/physiology , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Animals , CD40 Antigens/immunology , CD40 Antigens/metabolism , Humans , Interleukin-6/biosynthesis , Multiple Myeloma/etiology
13.
Clin Cancer Res ; 2(3): 531-9, 1996 Mar.
Article in English | MEDLINE | ID: mdl-9816200

ABSTRACT

We previously showed that combined neoadjuvant doxorubicin (DOX) treatment and orthotopic liver transplantation produced a 3-year tumor-free survival rate of 54% in stage II-IVa nonresectable hepatocellular carcinomas (HCCs). These patients received posttransplant immunosuppressive doses of cyclosporin A (CsA). CsA has been shown to modify the function of a membrane P-glycoprotein (Pgp) whose overexpression is associated with a multidrug-resistant (MDR1) phenotype. This study utilized HCC cell lines to characterize the in vitro chemomodulatory properties of CsA as found in posttransplant patient plasma to consider the hypothesis that CsA may prolong posttransplant survival by enhancing the therapeutic efficacy of DOX against multidrug-resistant hepatoma cells. We characterized Pgp expression in the HCC lines Hep3B, Hep G2, and SK-HEP-1 by immunohistochemistry and the reverse transcription-polymerase chain reaction. The combined cytotoxicity of DOX + CsA was examined by [3H]thymidine uptake and flow cytometric drug-retention assays. Pgp expression was assessed further after prolonged (10-day) treatment with CsA. Hep3B and Hep G2 cells expressed low to moderate levels of Pgp. The effective DOX dose required for inhibiting MDR1(+) Hep3B and Hep G2 cell proliferation by 50% (DOX IC50) was 44.5 ng/ml and 43.5 microgram/ml, as compared with 10.7 ng/ml for Pgp-negative SK-HEP-1 cells. Optimal concentrations of CsA (0.8 micrometer) lowered DOX IC50 for Hep3B cells and Hep G2 cells by 6-fold and 4-fold, respectively. Similarly, plasma from patients containing immunosuppressive levels of CsA lowered DOX IC50 of the MDR1(+) Hep G2 cells by up to 4-fold. Prolonged exposure to CsA did not affect its chemosensitizing capacity or Pgp expression of HCC cells. PSC-833, a nonimmunosuppressive analogue of CsA, was equally effective in reducing the DOX IC50 of MDR1(+) HCC cells. CsA and PSC-833 increased drug retention by approximately 75%, but did not significantly affect hepatoma cell viability or Pgp expression. Pharmacological concentrations of cyclosporin analogues, including one nonimmunosuppressive form, enhance DOX cytotoxicity of MDR1(+) HCC cells by modulating drug retention. CsA as found in posttransplant patient plasma enhanced DOX cytotoxicity to human MDR1(+) hepatoma cells in vitro, albeit at less than optimal chemosensitizing concentrations. Prolonged exposure to CsA did not affect its chemosensitizing properties or block Pgp expression of HCC cells. These findings support our hypothesis that in vivo immunosuppressive levels of CsA may enhance DOX chemotherapeutic efficacy on MDR1(+) HCC cells.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , Carcinoma, Hepatocellular/drug therapy , Cyclosporine/blood , Immunosuppressive Agents/blood , Liver Neoplasms/drug therapy , Liver Transplantation , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Cyclosporins/pharmacology , Doxorubicin/pharmacology , Humans
14.
Blood ; 84(9): 3026-33, 1994 Nov 01.
Article in English | MEDLINE | ID: mdl-7524765

ABSTRACT

Ligand binding of the B-cell lineage antigen CD40 enhances growth and interleukin-6 (IL-6) secretion in human B cells (the CD40/IL-6 loop). IL-6 has an autocrine and paracrine role in human multiple myeloma (MM) cell growth. With the use of the CD40 monoclonal antibody (MoAb) G28-5, we examined CD40 expression and the effect of CD40 binding on MM clonogenic colony (MCC) formation to characterize the IL-6/CD40 loop activity in MM. CD40 was expressed on plasmacytoid cells in 21 of 28 plasma cell dyscrasia (PCD) bone marrow (BM) biopsies tested (10 of 14 MM, 2 of 2 Waldenstrom's macroglobulinemia [WM], 2 of 2 plasma cell leukemia [PCL], 6 of 8 monoclonal gammopathy of undetermined significance [MGUS], and 1 of 2 primary amyloidosis [AL]). G28-5 binding increased MCCs by 35% to 150% in 11 of 17 CD40+ PCD BM cultures, but did not affect MCC formation in CD40- specimens or normal BM colony forming units (CFU-GEMM, CFU-GM, BFU-E). Responsive cultures originated from BM of patients with MM (2 of 5 cases tested), WM (2 of 2), PCL (2 of 2), and MGUS (5 of 6). CD40-responsiveness was not significantly inhibited by the presence of an anti-IL-6 MoAb (2 of 2 MGUS cultures tested), and did not correlate with the capacity to respond to IL-6 stimulation (n = 17, P > .05) or a detectable level of endogenous IL-6 (n = 15, P > .05). Additional studies were performed with PCD cell lines to characterize the interrelationship of CD40 activation and IL-6 production. Fifty percent to greater than 95% of cells from the RPMI 8226 and ARH77 lines expressed CD40, whereas 6% of U266 cells were CD40+. For RPMI 8226, ARH-77, and U266 cells, the increased MCC formation after anti-CD40 stimulation was not affected by the presence of an anti-IL-6 neutralizing MoAb and was not accompanied by detectable IL-6 secretion. There was no apparent increase in IL-6 mRNA transcription following G28-5 treatment of U266 or RPMI 8226 cells. Our observations indicate that CD40 is expressed in a subset of human myeloma cells present in various PCDs. Cell-line studies suggest that the CD40+ myeloma cell may regulate MM clonogenic colony formation without activating the IL-6 pathway.


Subject(s)
Antigens, CD/immunology , Antigens, Differentiation, B-Lymphocyte/immunology , Multiple Myeloma/pathology , Paraproteinemias/pathology , Antigen-Antibody Reactions , Bone Marrow/immunology , Bone Marrow/pathology , CD40 Antigens , Clone Cells , Gene Expression , Humans , Interleukin-6/physiology , Multiple Myeloma/immunology , Paraproteinemias/immunology , Plasma Cells/immunology , Plasma Cells/pathology , RNA, Messenger/genetics , Tumor Cells, Cultured
15.
Anticancer Res ; 13(3): 593-7, 1993.
Article in English | MEDLINE | ID: mdl-8391243

ABSTRACT

Human multiple myeloma (MM) xenografts have been difficult to establish in athymic mice. We examined the feasibility of establishing human MM xenograft growth in SCID mice following subcutaneous (sc) injection of 1-2 x 10(7) cells from the human plasma cell dyscrasia (PCD) cell lines RPMI 8226 and ARH-77. SC tumors emerged in 67% (6/9) of RPMI 8226- and 6 of 6 ARH-77-injected mice after a latency period of 9-54 days, and reached 19-35 mm in diameter before the mice were sacrificed. RPMI 8226 and ARH-77 primary tumor DNA hybridized positively with the human genome probe Alul-(Blur8), confirming successful engraftment of the human MM cell lines. The RPMI 8226 xenografts comprised predominantly of plasmacytoid cells that expressed the relevant cytoplasmic immunoglobulin (cIg) light chain isotype. Xenografted RPMI 8226 cells also expressed CD10 (CALLA; 44% reactive cells), CD38 (OKTIO; 69%), CD5 (49%), and reacted with the MM monoclonal antibody MM4 (39%). Human MM growth appeared to be localized subcutaneously for both RPMI 8226 and ARH-77 xenografts. There were no detectable metastatic foci in kidney, brain, heart, or bone marrow. Whereas diffuse plasma cell infiltrates were observed in spleen, GI tract, and lung biopsies of tumor-bearing mice, these infiltrates were of host origin according to immunophenotyping and DNA analyses. Neither the originating RPMI 8226 line nor its SCID mouse xenograft expressed Epstein Barr virus (EBV) genome sequences. These observations indicate that both EBV- (RPMI 8226) and EBV+ (ARH-77) cell lines can be successfully propagated in SCID mice.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Antigens, Neoplasm/analysis , Multiple Myeloma , Myeloma Proteins/analysis , Neoplasm Transplantation , Transplantation, Heterologous , Animals , Antibodies, Monoclonal , Cell Line, Transformed , Feasibility Studies , Genome, Human , Genome, Viral , Herpesvirus 4, Human/genetics , Humans , Mice , Mice, SCID , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Tumor Cells, Cultured/immunology , Tumor Cells, Cultured/pathology
16.
Cancer Res ; 53(6): 1392-6, 1993 Mar 15.
Article in English | MEDLINE | ID: mdl-8443818

ABSTRACT

We have successfully engrafted a human multiple myeloma cell line, ARH-77, into C.B. 17 SCID mice. When ARH-77 cells were injected s.c., tumors grew only at the site of inoculation (five of five). When ARH-77 cells were injected i.v. tumors did not grow in any of the mice (zero of five). However, when mice were given gamma-irradiation with 150 rads and then inoculated i.v. with 10(7) ARH-77 cells, 100% (13 of 13) of the mice developed tumors. Hind leg paralysis was observed in 13 of 16 mice as a result of compression of the spinal cord by tumor. Histological analysis demonstrated that myeloma cells proliferated and formed osteolytic lesions (15 of 16) in the vertebrae and bones of the skull (14 of 16). Tumor cells also invaded the brain and meninges (14 of 16), lung (13 of 15), liver (seven of 15), and kidney (two of 15). Flow cytometric analysis demonstrated that the phenotype of 31% of the bone marrow cells in the vertebrae and 79% of s.c. tumor cells was similar to ARH-77 cells (CD38+, PCA-1+, HLA-Classes 1 and II+). Furthermore, DNA hybridization with a human AluI probe confirmed their human origin. ARH-77-derived human immunoglobulin was detected in the serum of SCID/ARH-77 mice by ELISA. These observations demonstrate systemic involvement of human multiple myeloma following i.v. injection of ARH-77 cells into irradiated mice. This in vivo model should be useful for evaluating new therapeutic modalities for myeloma.


Subject(s)
Disease Models, Animal , Multiple Myeloma/pathology , Transplantation, Heterologous , Animals , Flow Cytometry , Humans , Immunoglobulins/blood , Mice , Mice, SCID , Neoplasm Transplantation , Nucleic Acid Hybridization , Phenotype , Tumor Cells, Cultured
17.
Prog Clin Biol Res ; 333: 155-64, 1990.
Article in English | MEDLINE | ID: mdl-2308979

ABSTRACT

We examined the effectiveness of a previously characterized plasma cell-reactive monoclonal antibody (MoAb), MM4, in eliminating multi-drug resistant (MDR1) multiple myeloma (MM) clonogenic colony-forming cells (CCCs). MDR1 sublines with 6-fold (RPMI8226/DOX6) and 40-fold (RPMI 8226/DOX40) resistance to doxorubicin (DOX) were selected from the chemosensitive MM parent line RPMI 8226/S. Both sublines remained reactive with plasma cell MoAbs MM4 and PCA-1, as measured by flow cytometric immunophenotype analysis. MM4 and rabbit complement (C') were cytotoxic to MDR DOX6 (74 +/- 8.5%) and DOX40 (75 +/- 11.3%) cells as well as to chemosensitive 8226/S (80 +/- 5.6%) cells. Treatment with MM4 + C' depleted up to 3 logs of chemosensitive and MDR myeloma CCCs (8226/S: 99.26 +/- 0.52%; DOX6 99.91 +/- 0.08%' DOX40 99.15 +/- 0.55%). In addition, this approach abrogated the selfrenewing capacity of chemoresistant and MDR1 myeloma cell lines, according to doubling time analyses. By comparison, the P-glycoprotein-reactive MoAb MRK-16 and C' was effective in deleting MDR1 CCCs (DOX10: 95.71 +/- 2.51%; DOX40: 99.61 +/- 0.43%) but affected chemosensitive myeloma CCCs only slightly (5.93 +/- 14.52%). When DOX40 cells were mixed with normal bone marrow (BM) in a ratio of 10:90 (MM:BM), treatment with MM4 plus C' deleted MM CCCs (98.80 +/- 0.71%) without affecting the majority of normal BM progenitors. The combination of MM4 and MRK-16 did not enhance MDR myeloma CCC depletion. These observations suggest that MM4 + C' may be useful for depleting MDR as well as chemosensitive myeloma clonogenic cells from human bone marrow.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Bone Marrow Cells , Complement System Proteins/therapeutic use , Multiple Myeloma/therapy , Neoplastic Stem Cells/immunology , Bone Marrow/immunology , Cell Survival/immunology , Drug Resistance , Humans , Multiple Myeloma/immunology , Phenotype , Tumor Cells, Cultured
18.
Cancer Res ; 49(17): 4829-34, 1989 Sep 01.
Article in English | MEDLINE | ID: mdl-2569359

ABSTRACT

Patients with multiple myeloma (MM) commonly become refractory to chemotherapy despite a favorable response to induction treatment. We examined the effectiveness of a previously characterized plasma cell-reactive monoclonal antibody, MM4, in eliminating MM clonogenic colony-forming cells (CCC) with a multidrug-resistant (MDR) phenotype. Experiments were performed using MM cell lines that exhibit 6 (RPMI 8226/DOX6)- and 40 (RPMI 8226/DOX40)-fold resistance to doxorubicin (DOX). Both lines were selected from the chemosensitive MM line RPMI 8226/S and were cross-resistant to mitoxantrone, acronycine, etoposide, and vincristine. Surface marker analysis conducted in this study showed that DOX6 and DOX40 overexpressed the MDR1 gene product p170. Both MDR lines remained reactive to the plasma cell-reactive monoclonal antibodies MM4 and PCA-1 and expressed the relevant cytoplasmic immunoglobulin light chain. Treatment with MM4 and rabbit complement (C') was equally cytotoxic to RPMI 8226/S [80 +/- 5.6% (SD)], DOX6 [74 +/- 8.5], and DOX40 cells [75 +/- 11.3%], based on short-term chromium release studies. Furthermore, MM4 + C' deleted up to 3 logs of CCC colonies from chemosensitive and MDR lines (RPMI 8226/S, 99.87 +/- 0.11%; DOX6, 99.91 +/- 0.08%; DOX40, 99.55 +/- 0.44%). By comparison, the P-glycoprotein-reactive monoclonal antibody MRK-16 and C' inhibited tumor colony formation of MDR cells (8226/DOX6, 95.71 +/- 2.51%; 8226/DOX40, 99.61 +/- 0.43%) but affected that of chemosensitive cells only slightly (8.9 +/- 17.8%). In an attempt to optimize the depletion of myeloma CCC, MM4 was used together with MRK-16. This approach resulted in uniform depletion of myeloma clonogenic colony-forming cells from the chemosensitive (98.32 +/- 1.53%, n = 4) and MDR lines (8226/DOX6, 98.83 +/- 0.08%, n = 4; 8226/DOX40 99.29 +/- 0.62, n = 7) but did not result in enhanced CCC depletion. When DOX40 cells were mixed with normal bone marrow (BM) in the ratio of 90:10 (BM:MM), either MM4 or MRK-16 and C' depleted MM colonies (98.8 +/- 0.71% and 98.10 +/- 1.0%, respectively) without affecting the majority of BM progenitor cells. These observations suggest that either MM4 or MRK-16 is useful for depleting MDR myeloma clonogenic colony-forming cells.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Membrane Glycoproteins/immunology , Multiple Myeloma/therapy , Plasma Cells/immunology , ATP Binding Cassette Transporter, Subfamily B, Member 1 , Antineoplastic Agents/pharmacology , Bone Marrow Cells , Doxorubicin/pharmacology , Drug Resistance , Humans , Multiple Myeloma/immunology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/immunology , Phenotype , Tumor Cells, Cultured
19.
Cancer Res ; 49(15): 4103-8, 1989 Aug 01.
Article in English | MEDLINE | ID: mdl-2545333

ABSTRACT

This study examined the lymphokine-activated killer (LAK) cell cytotoxicity on monoclonal antibody (MoAb)-bound tumor cells from the human small cell lung carcinoma cell lines H69 and H128. LAK cells were generated from normal peripheral blood mononuclear cells by incubation with interleukin 2 for 3 or more days. Cells from the LAK culture were cytotoxic to natural killer-sensitive (K562, 84% cytotoxicity) and natural killer-resistant (Daudi, 85%; H69 and H128, 69% and 97%, respectively) cell lines, and to freshly excised human lung (49%) and breast (57%) tumors. LAK cytotoxicity to H69 or H128 cells was significantly augmented by target cell preincubation with the small cell lung carcinoma-reactive MoAbs 1096 (increases of up to 271%) or 5023 (up to 223%). SCLC 5023 or 1096 did not enhance LAK cytotoxicity to Daudi cells of lymphoblastoid origin. Pretreatment of LAK cells with an anti-Fc receptor antibody blocked MoAb augmentation by 1096 or 5023 (but not LAK cytotoxicity), suggesting that LAK-MoAb interaction may be mediated by Fc binding. LAK activity coincided with emergence of a large cell [interleukin 2-stimulated large mononuclear leukocyte (LML)] subset expressing the CD16 and NKH-1 surface determinants. Serial immunophenotyping of the LAK cell culture harvested at Days 3, 5, and 7 indicated that the level of LAK cytotoxicity, with or without MoAb augmentation, correlated with frequency of NKH-1-reactive LMLs. These observations support the hypothesis that LAK cytotoxicity is mediated by a NKH-1-reactive LML subpopulation. Antitumor cytotoxicity may be augmented by tumor-reactive MoAbs through Fc binding to this LML subset.


Subject(s)
Antibodies, Monoclonal/immunology , Carcinoma, Small Cell/immunology , Interleukin-2/pharmacology , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Antigens, Surface/analysis , Cytotoxicity, Immunologic , Humans , Immunotherapy , Receptors, Fc/physiology , Recombinant Proteins/pharmacology
20.
Cancer Detect Prev ; 12(1-6): 621-35, 1988.
Article in English | MEDLINE | ID: mdl-3180147

ABSTRACT

The monoclonal antibody MM4 reacts with human myeloma cells from plasma cell dyscrasia (PCD)-derived cell lines and bone marrow (BM) biopsies from PCD patients, but not with normal BM or peripheral blood mononuclear (PBM) cells. We examined cytotoxicity of MM4 and rabbit complement (MM4:C') on mixtures of normal BM mononuclear cells and myeloma cells from three different PCD-derived cell lines, RPMI 8226, GM 1312, or ARH-77. For cell preparations containing 10% myeloma cells, treatment with MM4 (500 micrograms per 10(5) cells, 4 degrees C, 60 min) and two cycles of complement (1:8, 23 degrees C, 2 x 30 min) consistently eliminated 2 logs or more of clonogenic myeloma stem cells, as determined by colony growth assays and limiting dilution analysis (99.4%, 98.9%, and 99.96% reduction of RPMI 8226, GM 1312, and ARH-77 cells, respectively). The majority of normal marrow progenitors were spared (inhibition of CFU-C: 10-13%; BFU-E: 0%). These observations suggest that MM4 may be useful for selective depletion of human myeloma clonogenic stem cells from bone marrow ex vivo.


Subject(s)
Antibodies, Monoclonal/immunology , Bone Marrow/pathology , Cell Separation/methods , Complement System Proteins , Multiple Myeloma/pathology , Neoplastic Stem Cells , Algorithms , Animals , Antigen-Antibody Reactions , Bone Marrow/immunology , Cell Line , Cytotoxicity, Immunologic , Hematopoietic Stem Cells/immunology , Humans , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL
...