Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where James D. Liddil is active.

Publication


Featured researches published by James D. Liddil.


Biochemical Pharmacology | 1987

Mitomycin C resistant L1210 leukemia cells: Association with pleiotropic drug resistance

Robert T. Dorr; James D. Liddil; Jeffrey M. Trent; William S. Dalton

A mitomycin C-resistant (MMCR) strain of L1210 mouse leukemia was developed by continuous drug exposure in vitro. MMC concentrations were increased in a stepwise fashion beginning at 0.033 microM and ending at 0.34 microM. This produced a 10-fold resistant cell line over the parental line. Resistance simultaneously developed to anthracene and anthracycline DNA intercalators, to vinca alkaloids and epipodophyllotoxins but not to cisplatin, bleomycin, fluorouracil or ionizing X-rays. MMC resistance was reversed using the membrane-active agent verapamil. The level of non-protein sulfhydryls was increased 2-fold in the MMCR cells. Intracellular uptake of unchanged MMC was reduced by 40% in the MMCR cells. Cytogenetic analyses demonstrated no recognizable clonal chromosomal alterations unique to the resistant subline and no evidence of double minutes or homogeneously staining regions in the DNA. Gel renaturation analysis failed to document the presence of an amplified DNA domain. Southern blotting of parental and MMCR DNA using a cDNA probe (CHP1) for the P-glycoprotein gene also failed to demonstrate amplification or rearrangement of P-glycoprotein-related homologous sequences. However, an Mr 180,000 glycoprotein was detected in the plasma membranes from MMCR cells. This protein also specifically reacted with a monoclonal antibody (C219) to the P-glycoprotein of Ling and co-workers [Kartner et al., Nature, Lond. 316, 820 (1985)]. These results suggest a pleiotropic drug resistance pattern in the MMCR cells, associated with membrane glycoprotein alterations, enhanced non-protein sulfhydryl levels, and reduced MMC accumulation. This is a novel observation for a resistant cell line selected with an alkylating agent.


Investigational New Drugs | 1986

Cytotoxic effects of glutathione synthesis inhibition by L-buthionine-(SR)-sulfoximine on human and murine tumor cells

Robert T. Dorr; James D. Liddil; Michelle J. Soble

The glutathione (GSH) synthesis inhibitor, buthionine sulfoximine (BSO) was tested for cytotoxicity and thiol depletion in murine and human tumor cells in vitro, and for its antitumor activity and toxicity in vivo. The cell lines used in these studies included murine L-1210 leukemia, human RPMI 8226 myeloma, MCF-7 breast cancer and WiDr colon carcinoma. Soft agar colony forming assays showed that BSO was most effective at reducing tumor colony formation when exposed continuously to cells in vitro. Drug concentrations which inhibited colony formation to 50% of control levels ranged from 2.0–6.2 mM (for 1 hour exposures), 2–100 mM for 24 hour exposures and 0.4–1.40 μM (for continuous BSO exposures). Human myeloma cells proved most sensitive to BSO. In vitro cytotoxicity correlated with depletion of intracellular nonprotein sulfhydryls to ≤ 10% of control values in both L-1210 and 8226 cells. This was routinely achieved with prolonged exposures to mM BSO concentrations for > 24 hours. Normal mice tolerated high BSO doses (up to 5.0 g/kg) without evidence of acute toxicity. BSO was not active against L-1210 leukemia-bearing DBA/2 mice. When tested in vivo against MOPC-315 plasmacytoma-bearing BALB/c mice, BSO was not active at doses up to 4.0 g/kg. In contrast, the bifunctional alkylating agent melphalan (L-PAM) was active against MOPC-315 and this activity was enhanced by a 24 hour pretreatment of mice with 50 mg/kg of L-BSO. This BSO dose was shown to significantly reduce sulfhydryl levels in the liver (50% of control) and kidney (20% of control) but not in the bone marrow (100% of control). The enhancement by BSO was most significant only for the lower doses of L-PAM tested. These results suggest that BSO may not have direct antitumor activity, but that it can enhance cytotoxicity from a classic alkylating agent in vivo. Due to its low toxicity, BSO should be tested in combination with either anticancer agents which are dependent on (GSH) for detoxification and potential drug resistance.


Cancer Investigation | 1993

The Effect of Anticancer Drug Sequence in Experimental Combination Chemotherapy

Alfonso L. Adel; Robert T. Dorr; James D. Liddil

A series of drug combination sequence studies was conducted in vitro using HEC-1A human endometrial carcinoma cells or 8226 myeloma cells. Four drugs were evaluated for schedule-dependent and sequence-dependent inhibition of human tumor colony formation in soft agar. Six different two-drug combinations were analyzed using the median dose effect method, and three different three-drug combinations were examined using the cumulative surviving fraction method. The results show that the specific sequence and method of drug exposure significantly influenced the production of antagonistic, additive, or synergistic cytotoxicity patterns. Drug combinations that were consistently synergistic included bleomycin or mitomycin C and cisplatin in 8226 cells, and etoposide plus bleomycin in human endometrial cancer (HEC-1A) cells. Most other two-drug combinations of bleomycin, etoposide, cisplatin, and mitomycin C were antagonistic in vitro, irrespective of the sequence of exposure. Among the three-drug combinations tested, consistent synergism was noted with cisplatin, etoposide, and bleomycin when either of the latter two agents was tested as a continuous exposure in vitro. Within individual two- and three-drug combinations, it was possible to observe synergism, additivity, or antagonism based on the particular exposure sequence tested. These results suggest that antitumor agent cytotoxicity in vitro can be radically influenced by the sequence of drug administration, a feature commonly overlooked in many clinical combination drug regimens.


Investigational New Drugs | 1995

Preclinical pharmacokinetics and antitumor activity of imexon

Robert T. Dorr; James D. Liddil; Mary Kay Klein; Evan M. Hersh

SummaryImexon is an aziridine compound originally studied for immune-enhancing effects on lymphocytes. The drug was well-tolerated in humans and was shown to be active in a variety of animal tumor models. Recently, imexon has demonstrated antitumor activity in human multiple myeloma cell linesin vitro. The pharmacokinetics of the compound using a normal phase HPLC assay were studied in normal mice and in dogs with mast cell tumors. Doses of 100 mg/kg given intraperitoneally produced peak plasma levels over 100 (μml in mice and the drug was rapidly eliminated with half lives of 8 minutes (α phase) and 29 minutes (β phase). Only 20% of an oral imexon dose was absorbed in the mouse. In dogs, the α and β phase half lives ranged from 18–26 minutes and 91–110 minutes, respectively. Peak levels over 100 μg/ml were obtained following intravenous doses of 12.5 mg/kg and 25 mg/kg. Imexon was active in mice bearing either P-388 or L-1210 leukemia, but not in mice with B-16 melanoma. These results suggest that cytotoxic drug concentrations can be obtainedin vivo and that imexon is active in lymphoproliferative tumors.


Journal of Clinical Oncology | 1986

Mitomycin C skin toxicity studies in mice: reduced ulceration and altered pharmacokinetics with topical dimethyl sulfoxide.

Robert T. Dorr; Michelle J. Soble; James D. Liddil; James H. Keller

A series of toxicologic and pharmacokinetic studies were performed in BALB/c mice administered intradermal (ID) mitomycin C (MMC) at doses of .015 to 0.25 mg. Dose-dependent skin ulcers were produced at clinically relevant MMC dose levels of .05 and .075 mg (3.6 to 10.7 mg/m2). These doses produced peak ulcers of 0.15 to 0.22 cm2, respectively, one to five days after injection. The integrated ulcer area X time values (area under the curve [AUC] ulceration) were 0.89 and 3.11 cm2 X d. A large number of local pharmacologic adjuvants were found to be ineffective at reducing MMC ulceration after proximal ID injection. These included diphenhydramine, catalase, heparin, hyaluronidase, hydrocortisone, cysteine, N-acetylcysteine, lidocaine, vitamin E, and superoxide dismutase. Also, neither topical heating nor cooling of skin reduced MMC ulcerations. In contrast, a single topical application of a 100% dimethyl sulfoxide (DMSO) solution completely prevented 0.025 mg MMC-induced skin ulceration and significantly reduced .075 mg MMC ulceration (P less than .05 by multiple range tests). Topical DMSO also altered the disposition of ID MMC in mouse skin but not in plasma. Unexpectedly, the DMSO applications slowed MMC elimination from the skin. DMSO significantly increased the AUC for MMC in skin from 0.89 to 2.25 ng/h/mL of tissue (P less than .05). DMSO did not alter the degree of protein binding in skin tissue nor the in vitro chemical stability of MMC in skin tissue homogenates. These results show that experimental MMC-induced skin ulcers in mice can be ameliorated with an immediate application of topical DMSO. This effect is not due to enhanced systemic drug uptake, but may be due to reduced reactivity of MMC with target cellular nucleophiles.


Anti-Cancer Drugs | 2001

Preclinical antitumor activity of the azonafide series of anthracene-based DNA intercalators

Robert T. Dorr; James D. Liddil; Salah M. Sami; William A. Remers; Evan M. Hersh; David S. Alberts

The azonafides are a series of anthracene-based DNA intercalators which inhibit tumor cell growth in vitro at low nanomolar concentrations and are not affected by the multidrug resistance phenomenon (MDR). Prior studies have described antitumor efficacy in murine tumor models including L-1210 and P-388 leukemias, and B-16 melanoma. The current results extend these cell line observations to human tumors tested in the NCI panel of 56 cell lines, in freshly isolated tumors tested in colony-forming assays in soft agar and in several animal models. In the NCI panel, the overall mean 50% cell kill (LC50) for the unsubstituted azonafide, AMP-1, was 10−5.53 M, with some selectivity noted in melanomas (10−6.22 M). The mean LC50 for the 6-ethoxy substituted analog, AMP-53, was 10−5.53 M, with some selectivity found in non-small cell lung cancer (10−5.91) and renal cell carcinoma (10−5.84). In freshly isolated human tumors tested in soft agar, there was marked activity (mean IC50 in μg/ml) for AMP-53 in four cell types: breast cancer (0.09), lung cancer (0.06), renal cell carcinomas (0.06) and multiple myeloma (0.03). These effects were superior to doxorubicin and to several other azonafides, including AMP-1, AMP-104 and the 6-hydroxyethoxy derivative, AMP-115. Compound AMP-1 was shown to be superior to amonafide in the mammary 16C breast cancer model in B6CF31 mice, but it had little activity in Colon-38 nor in M5076 ovarian sarcomas in vivo. Nine azonafides were evaluated in the Lewis lung cancer model in C57/bl mice, but only AMP-53 demonstrated significant efficacy with a treated/control×100% (T/C) value of 30%. Because AMP-53 demonstrated the greatest breadth of activity, it was then evaluated in several human tumor cell lines growing in mice with severe combined immunodeficiency disease (SCID). Only three tumors were sensitive (T/C<42%), including HL-60 leukemia (T/C=39%), MCF-7 breast cancer (T/C=39%) and A549 non-small cell lung cancer (T/C=37%). Overall, these results demonstrate that the 6-ethoxy substituted azonafide, AMP-53, has consistent (in vitro and in vivo) experimental antitumor activity in human breast and lung cancer, and could be considered for clinical testing in patients with MDR tumors.


Cancer Chemotherapy and Pharmacology | 1992

Cardiotoxicity of mitomycin A, mitomycin C, and seven N7 analogs in vitro.

Robert T. Dorr; Nancy G. Shipp; James D. Liddil; Bhashyam S. Iyengar; Kenneth R. Kunz; William A. Remers

SummaryThe alkylating antitumor agents mitomycin A (MMA), mitomycin C (MMC), and sevenN7 analogs were compared in terms of their cardiotoxic and antitumor activity in vitro. Neonatal rat-heart myocytes were sensitive to five of the compounds studied, including MMA, 7-dimethylamidinomitosane (BMY-25282), 7-(N-methyl-piperazinyl)-mitosane (RR-194),N7-(4-iodophenyl)-MMC (RR-208), andN7-(4-hydroxyphenyl)-MMC (M-83) in order of descending molar potency. MMA and RR-208 possessed the greatest cytotoxic potency against 8226 human myeloma tumor cells in vitro. Two of the nine mitomycins studied, BMY-25282 and M-83, showed greater cytotoxic potency for heart cells. For these two agents, the ratio of the 50% inhibitory concentration in heart cells to that in 8226 myeloma cells was 50 and 32, respectively. For the other analogs, the tumor-cell cytotoxic potency was much higher (ranging from 200 to 7,000). For the nine mitomycin compounds, a correlation was found between heart-cell toxicity and low reduction potentials (E1/2 values) ranging from −0.16 to −0.37 V. Thus, as the reduction potential decreased (easier reducibility), the cardiotoxic potency in vitro increased (r = 0.81). In contrast, mitomycins with reduction potentials of higher than −0.37 V were much less potent cardiotoxins. Thus, mitomycin C (E1/2 = −0.45 V) was noncardiotoxic even when tested at concentrations 100-fold above those pharmacologically achievable in humans. Mitomycin C also failed to enhance doxorubicin (Adriamycin) cardiotoxicity in vitro. Importantly, no correlation was found between the reduction potential and the antitumor activity of the nine analogs (n = 0.51), in this small series.


Cancer Chemotherapy and Pharmacology | 1991

Modulation of mitomycin C-induced multidrug resistance in vitro

Robert T. Dorr; James D. Liddil

SummaryA series of in vitro cytotoxicity studies were performed to achieve pharmacologic reversal of resistance to the alkylating agent mitomycin (MMC) in L-1210 leukemia cells. A multidrug-resistant (MDR), P-glycoproteinpositive cell line, RL-1210/.1 [11], was exposed to potential MDR modulators in the absence or presence of MMC. The following compounds did not reverse MMC-induced MDR: quinine, quinidine, lidocaine, procaine, dimethylsulfoxide (DMSO), dexamethasone, hydrocortisone, prednisolone, estradiol, and testosterone. Three agents were capable of reversing MMC resistance: progesterone, cyclosporin A, and verapamil. The R- and S-isomers of verapamil were equipotent, although they showed a 10-fold difference in cardiovascular potency (S>R). Some agents produced cytotoxic effects in MDR cells in the absence of MMC, including progesterone, quinine, and quinidine. The results suggest that R-verapamil and progesterone may have clinical utility in reversing MMC resistance in human tumors. Progesterone may be uniquely efficacious due to (a) its low toxicity in normal cells, (b) its selective cytotoxicity in MDR cells (in the absence of MMC), and (c) its ability to reverse MMC resistance in vitro. The findings also suggest that the P-glycoprotein induced by MMC differs from that induced by doxorubicin, which is highly sensitive to modulation by lysosomotropic amines such as quinine and quinidine.


Cancer Chemotherapy and Pharmacology | 1998

Analytical and biological inequivalence of two commercial formulations of the antitumor agent bleomycin

Robert T. Dorr; Ross O. Meyers; Kristi Snead; James D. Liddil

Abstract Bleomycin is an antitumor agent which is a mixture of glycopeptides containing at least 55–75% bleomycin A2 and 25–32% bleomycin B2 fractional composition. Two bleomycin formulations, bleomycin sulfate, USP (Blenoxane, Bristol-Myers Squibb Oncology, Princeton, N.J.) and bleomycin HCl (Tianjin Hebei Pharmaceutical, Tianjin, China) were compared analytically and biologically. Reverse-phase high-performance liquid chromatography (HPLC) analyses using the USP methodology showed that Blenoxane contained primarily (69%) bleomycin A2 and 29.3% bleomycin B2. In contrast, Tianjin-supplied bleomycin HCl contained 97% bleomycin A5 fraction. In vitro tumor cell growth inhibition assays showed equivalent activity in human OVCAR-3 ovarian cancer cells and slightly greater potency in murine L-1210 leukemia cells for the Tianjin formulation. In C57/Bl mice bearing B-16 melanoma tumors, Tianjin-supplied bleomycin produced slightly greater tumor growth inhibition at the expense of greater drug-induced lethality at higher dose levels. These studies show there are significant differences in two international bleomycin formulations. These compositional differences lead to altered biologic effects.


Nucleosides, Nucleotides & Nucleic Acids | 1994

Biologic Activity of a Nucleotide Conjugate Between Mitomycin C and Cytarabine Monophosphate

Robert T. Dorr; James D. Liddil; Bhashyam S. Iyengar; A. F. List; William A. Remers

Abstract A dual prodrug conjugate between the antimetabolite cytarabine monophosphate and the alkylating agent 2,7-diaminomitosene (derived from mitomycin C), cytaramycin, was synthesized and tested for antileukemic activity in sensitive and resistant tumors. The compound was active against parental L-1210, CCRF-CEM, HL-60 and K-562 leukemia cells but did not overcome resistance in sublines developed for (1) multidrug resistance (L-1210/MDR and K-562-R) or (2) for cytarabine resistance (CCRF-CEM/ARA-C and HL-60/ARA-C). Alkaline DNA elution tests demonstrate a predominance of strand breaking activity due to the cytarabine moiety, and a lesser degree of DNA crosslinking, due to the mitosene moiety. The conjugate was active in mice bearing P-388 leukemia (80% increased lifespan), but was not more effective than mitomycin C alone in mice bearing a cytarabine-resistant L-1210 cell line (38% to 31% increased lifespan). These findings suggest that mitomycin nucleotide conjugates do not overcome resistance to the...

Collaboration


Dive into the James D. Liddil's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jeffrey M. Trent

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge