Network


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

Hotspot


Dive into the research topics where Lai-Ming Ching is active.

Publication


Featured researches published by Lai-Ming Ching.


Journal of Experimental Medicine | 2007

The chemotherapeutic agent DMXAA potently and specifically activates the TBK1–IRF-3 signaling axis

Zachary J. Roberts; Nadege Goutagny; Pin Yu Perera; Hiroki Kato; Himanshu Kumar; Taro Kawai; Shizuo Akira; Ram Savan; David Van Echo; Katherine A. Fitzgerald; Howard A. Young; Lai-Ming Ching; Stefanie N. Vogel

Vascular disrupting agents (VDAs) represent a novel approach to the treatment of cancer, resulting in the collapse of tumor vasculature and tumor death. 5,6-dimethylxanthenone-4-acetic acid (DMXAA) is a VDA currently in advanced phase II clinical trials, yet its precise mechanism of action is unknown despite extensive preclinical and clinical investigations. Our data demonstrate that DMXAA is a novel and specific activator of the TANK-binding kinase 1 (TBK1)–interferon (IFN) regulatory factor 3 (IRF-3) signaling pathway. DMXAA treatment of primary mouse macrophages resulted in robust IRF-3 activation and ∼750-fold increase in IFN-β mRNA, and in contrast to the potent Toll-like receptor 4 (TLR4) agonist lipopolysaccharide (LPS), signaling was independent of mitogen-activated protein kinase (MAPK) activation and elicited minimal nuclear factor κB–dependent gene expression. DMXAA-induced signaling was critically dependent on the IRF-3 kinase, TBK1, and IRF-3 but was myeloid differentiation factor 88–, Toll–interleukin 1 receptor domain–containing adaptor inducing IFN-β–, IFN promoter-stimulator 1–, and inhibitor of κB kinase–independent, thus excluding all known TLRs and cytosolic helicase receptors. DMXAA pretreatment of mouse macrophages induced a state of tolerance to LPS and vice versa. In contrast to LPS stimulation, DMXAA-induced IRF-3 dimerization and IFN-β expression were inhibited by salicylic acid. These findings detail a novel pathway for TBK1-mediated IRF-3 activation and provide new insights into the mechanism of this new class of chemotherapeutic drugs.


British Journal of Cancer | 2002

Induction of endothelial cell apoptosis by the antivascular agent 5,6-Dimethylxanthenone-4-acetic acid.

Lai-Ming Ching; Cao Z; Kieda C; Zwain S; Jameson Mb; Bruce C. Baguley

5,6-Dimethylxanthenone-4-acetic acid, synthesised in this laboratory, reduces tumour blood flow, both in mice and in patients on Phase I trial. We used TUNEL (TdT-mediated dUTP nick end labelling) assays to investigate whether apoptosis induction was involved in its antivascular effect. 5,6-Dimethylxanthenone-4-acetic acid induced dose-dependent apoptosis in vitro in HECPP murine endothelial cells in the absence of up-regulation of mRNA for tumour necrosis factor. Selective apoptosis of endothelial cells was detected in vivo in sections of Colon 38 tumours in mice within 30 min of administration of 5,6-Dimethylxanthenone-4-acetic acid (25 mg kg−1). TUNEL staining intensified with time and after 3 h, necrosis of adjacent tumour tissue was observed. Apoptosis of central vessels in splenic white pulp was also detected in tumour-bearing mice but not in mice without tumours. Apoptosis was not observed in liver tissue. No apoptosis was observed with the inactive analogue 8-methylxanthenone-4-acetic acid. Positive TUNEL staining of tumour vascular endothelium was evident in one patient in a Phase I clinical trial, from a breast tumour biopsy taken 3 and 24 h after infusion of 5,6-Dimethylxanthenone-4-acetic acid (3.1 mg m−2). Tumour necrosis and the production of tumour tumour necrosis factor were not observed. No apoptotic staining was seen in tumour biopsies taken from two other patients (doses of 3.7 and 4.9 mg m−2). We conclude that 5,6-Dimethylxanthenone-4-acetic acid can induce vascular endothelial cell apoptosis in some murine and human tumours. The action is rapid and appears to be independent of tumour necrosis factor induction.


Cancer Chemotherapy and Pharmacology | 1995

Induction of tumour necrosis factor-α by single and repeated doses of the antitumour agent 5,6-dimethylxanthenone-4-acetic acid

Martin Philpott; Bruce C. Baguley; Lai-Ming Ching

Abstract5,6-Dimethylxanthenone-4-acetic acid (DMXAA), a low-molecular-weight biological response modifier scheduled for clinical evaluation, induced synthesis of tumour necrosis factor-α (TNF-α) in serum of mice, with maximal activity being observed at 2–3 h after administration. At a dose of 27.5 mg/kg, DMXAA induced similar TNF-α concentrations as did flavone-8-acetic acid given at its maximum tolerated dose (MTD; 330 mg/kg), whereas 8-methylxanthenone-4-acetic acid, which has no antitumour activity, did not induce serum TNF-α at its MTD (440 mg/kg). The dependence of schedule on TNF-α induction was studied by giving DMXAA to mice in two doses of 27.5 mg/kg each separated by different intervals. An interval of 0 (i.e 55 mg/kg given in a single dose) produced a TNF-α concentration 9-fold that produced hy a single dose of 27.5 mg/kg. This dose, although higher than the MTD of 30 mg/kg, did not affect the health of mice at the time of assay (3 h). An interval of 1 day produced very low levels of serum TNF-α after the second injection. An interval of 3 days produced high levels of serum TNF-α after the second injection (9-fold that detected in mice receiving 27.5 mg/kg in a single dose) but no long-term toxicity, whereas an interval of 7 days produced an intermediate response. Thus, the first dose can either potentiate or suppress the TNF-α response to a second dose. Mice with advanced subcutaneous colon 38 tumours were treated either with a single dose of DMXAA (27.5 mg/kg) or with a divided dose (two doses of 27.5 mg/kg given 3 days apart). Both the cure rate and the tumour-growth delay were enhanced by the divided-dose schedule. The results are relevant to the design of clinical administration schedules of DMXAA and emphasise the importance of TNF-α induction in the antitumour response.


British Journal of Cancer | 2004

Relationship between tumour endothelial cell apoptosis and tumour blood flow shutdown following treatment with the antivascular agent DMXAA in mice.

Lai-Ming Ching; Zwain S; Bruce C. Baguley

5,6-Dimethylxanthenone-4-acetic acid (DMXAA) is currently undergoing clinical evaluation as an antivascular agent for the treatment of cancer. We have previously demonstrated that DMXAA induces apoptosis of vascular endothelial cells in murine tumour sections and in a breast carcinoma biopsy from one patient in a Phase I trial. We wished to determine the tissue selectivity of this effect and its relationship to induced blood flow changes. Mice with Colon 38 tumours were treated with DMXAA and tissues were examined for apoptosis by TdT-mediated dUTP nick-end labelling (TUNEL). Hoechst 33342 was used to stain functional vessels, with the loss of stained vessels used as a measure of tumour vascular collapse. Treatment with DMXAA at 25 mg kg−1, its maximum tolerated dose (MTD), showed, after 3 h, a 12-fold increase in TUNEL staining of tumour vascular endothelial cells. In contrast, tissue from the heart, brain, liver and spleen showed no increase. Induction of apoptosis in tumour tissue was both dose-dependent, observable at doses as low as 5 mg kg−1, and time-dependent. Apoptosis was significantly lower in Colon 38 tumours of mice, with a targeted disruption in the TNF gene (TNF−/−), or in the TNF receptor 1 gene (TNFR−/−), as compared with that in wild-type mice. Increasing the DMXAA dose to 50 mg kg−1 in these knockout mice raised tumour apoptosis to a level comparable to that induced in wild-type mice given DMXAA at the MTD. For all the data, a significant correlation (r=0.94; P<0.001) was found between logarithmic percentage apoptosis induction and the logarithmic density of Hoechst-stained vessels. These results suggest that blood flow inhibition caused by DMXAA is tumour tissue-specific and is a consequence of induction of apoptosis in tumour vascular endothelial cells.


European Journal of Cancer and Clinical Oncology | 1987

Induction of Natural Killer Cell Activity by the Antitumour Compound Flavone Acetic Acid (NSC 347 512)

Lai-Ming Ching; Bruce C. Baguley

Flavone acetic acid (FAA), an antitumour drug at present undergoing clinical trial for cancer treatment, has been found to activate natural killer cell activity in spleen cells of mice following in vivo treatment at doses of 45-330 mg/kg. The activity of FAA was measured using a 51Cr-release assay, employing the YAC-1 lymphoma line as a target cell population, and was found to be comparable to that of the interferon inducer polyinosinic acid-polycytidylic acid. The induction of activity was blocked by anti-asialo GM1 antibody. The time course of induction of activity by FAA was similar to that of the induction of haemorrhagic necrosis of the colon 38 tumour in mice, suggesting that natural killer cells or other components of the immune system may be involved in the action of FAA.


International Journal of Cancer | 2005

Mechanisms of tumor vascular shutdown induced by 5,6-dimethylxanthenone-4-acetic acid (DMXAA): Increased tumor vascular permeability

Liangli Zhao; Lai-Ming Ching; Philip Kestell; Lloyd R. Kelland; Bruce C. Baguley

The novel vascular targeting agent 5,6‐dimethylxanthenone‐4‐acetic acid (DMXAA) has completed phase 1 clinical trial and has shown tumor antivascular activity in both mice and humans. We have investigated its ability to change tumor vascular permeability, relating it to tumor vascular perfusion and other responses. The murine colon 38 adenocarcinoma was grown in C57Bl wild‐type mice and mice lacking expression of either tumor necrosis factor receptor‐1 (TNFR1−/−) or TNF (TNF−/−). Tumor vascular permeability, as measured by extravasation of albumin‐Evans Blue complexes 4 hr after DMXAA treatment, was significantly increased in tumor tissue in C57Bl, TNFR1−/− and TNF−/− mice but not in normal (skin) tissue. Significant linear relationships were found between increased tumor vascular permeability, decreased functioning tumor blood vessels (measured by Hoechst 33342 staining at 4 hr), increased plasma 5‐hydroxyindole‐3‐acetic acid concentrations (as a measure of serotonin release by platelets) and the degree of induced tumor hemorrhagic necrosis. The results support the hypothesis that DMXAA increases tumor vascular permeability both directly and through the induction of other vasoactive mediators, including TNF. DMXAA might be useful clinically to potentiate the vascular permeability of other anticancer modalities such as cytotoxic drugs, antibodies, drug conjugates and gene therapy.


British Journal of Cancer | 1995

Effect of thalidomide on tumour necrosis factor production and anti-tumour activity induced by 5,6-dimethylxanthenone-4-acetic acid

Lai-Ming Ching; Z. F. Xu; B. H. Gummer; Palmer Bd; W. R. Joseph; Bruce C. Baguley

The investigational anti-tumour agent, 5,6-dimethylxanthenone-4-acetic acid (5,6-MeXAA), an analogue of flavone acetic acid (FAA), has been scheduled for clinical evaluation. Like FAA, 5,6-MeXAA exhibits excellent experimental anti-tumour activity and is an efficient inducer of cytokines in mice. We have examined the effect of pharmacological suppression of tumour necrosis factor (TNF) production on the anti-tumour activity of 5,6-MeXAA, taking advantage of previous observations that TNF production in response to endotoxin in vitro is inhibited by thalidomide. Thalidomide at doses of between 8 and 250 mg kg-1 efficiently suppressed serum TNF activity in response to 5,6-MeXAA at its optimal TNF inducing dose of 55 mg kg-1. Suppression was achieved when thalidomide was administered at the same time as, or up to 4 h before, 5,6-MeXAA. Under conditions in which TNF activity was suppressed, the degree of tumour haemorrhagic necrosis and the proportion of cures in the subcutaneous Colon 38 tumour were increased. In mice administered thalidomide (100 mg kg-1) together with 5,6-MeXAA (30 mg kg-1), complete tumour regression was obtained in 100% of mice, as compared with 67% in mice receiving 5,6-MeXAA alone. The results suggest a possible new application for thalidomide and pose new questions about the action of 5,6-MeXAA and related compounds.


European Journal of Cancer and Clinical Oncology | 1988

Cell Line Selectivity and DNA Breakage Properties of the Antitumour Agent N-(2- (Dimethylamino) ethyl) acridine-4-carboxamide: Role of DNA Topoisomerase II*

Erasmus Schneider; Sandra J. Darkin; Penelope A. Lawson; Lai-Ming Ching; R.K. Ralph; Bruce C. Baguley

N-[2-(Dimethylamino)ethyl]acridine-4-carboxamide (NSC 601316) is a DNA intercalating experimental antitumour agent which is curative against the Lewis lung carcinoma in mice. Its action has been compared with amsacrine, its inactive isomer oAMSA, the solid tumour active derivative CI-921 (NSC 343499), a C-6 methylene chain-linked bisacridine (NSC 210733), 9-aminoacridine and quinacrine. All compounds inhibited the unknotting of phage P4 DNA by topoisomerase II in nuclear extracts prepared from L1210 cells. NSC 601316 inhibited growth of cultured L1210, P388, P/AMSA (P388 resistant to amsacrine) and P/ACTD (resistant to actinomycin D) cell lines at concentrations of 87, 150, 2020 and 150 nM respectively. A 1 h drug exposure to 0.85 microM NSC 601316 killed 50% of L1210 cells. L1210 cells treated for 1 h with NSC 601316 accumulated DNA breaks and protein-DNA cross-links. There was a good correlation between DNA breakage and cytotoxicity, but the relationship between drug concentration and number of protein-DNA cross-links was non-linear and differed from that of amsacrine and CI-921. There was also a positive correlation between the degree of cross-resistance of P/AMSA cells (which have altered topoisomerase II function) and ability to induce DNA breakage or protein-DNA complexes. The results suggest that topoisomerase II is the target of action of NSC 601316.


British Journal of Cancer | 2002

The antitumour activity of 5,6-dimethylxanthenone-4-acetic acid (DMXAA) in TNF receptor-1 knockout mice

Zhao L; Lai-Ming Ching; Kestell P; Bruce C. Baguley

5,6-dimethylxanthenone-4-acetic acid, a novel antivascular anticancer drug, has completed Phase I clinical trial. Its actions in mice include tumour necrosis factor induction, serotonin release, tumour blood flow inhibition, and the induction of tumour haemorrhagic necrosis and regression. We have used mice with a targeted disruption of the tumour necrosis factor receptor-1 gene as recipients for the colon 38 carcinoma to determine the role of tumour necrosis factor signalling in the action of 5,6-dimethylxanthenone-4-acetic acid. The pharmacokinetics of 5,6-dimethylxanthenone-4-acetic acid, as well as the degree of induced plasma and tissue tumour necrosis factor, were similar in tumour necrosis factor receptor-1−/− and wild-type mice. However, the maximum tolerated dose of 5,6-dimethylxanthenone-4-acetic acid was considerably higher in tumour necrosis factor receptor-1−/− mice (>100 mg kg−1) than in wild-type mice (27.5 mg kg−1). The antitumour activity of 5,6-dimethylxanthenone-4-acetic acid (25 mg kg−1) was strongly attenuated in tumour necrosis factor receptor-1−/− mice. However, the reduced toxicity in tumour necrosis factor receptor-1−/− mice allowed the demonstration that at a higher dose (50 mg kg−1), 5,6-dimethylxanthenone-4-acetic acid was curative and comparable in effect to that of a lower dose (25 mg kg−1) in wild-type mice. The 5,6-dimethylxanthenone-4-acetic acid -induced rise in plasma 5-hydroxyindoleacetic acid, used to reflect serotonin production in a vascular response, was larger in colon 38 tumour bearing than in non-tumour bearing tumour necrosis factor receptor-1−/− mice, but in each case the response was smaller than the corresponding response in wild-type mice. The results suggest an important role for tumour necrosis factor in mediating both the host toxicity and antitumour activity of 5,6-dimethylxanthenone-4-acetic acid, but also suggest that tumour necrosis factor can be replaced by other vasoactive factors in its antitumour action, an observation of relevance to current clinical studies.


The Journal of Pathology | 2009

A role for interleukin-6 in spreading endothelial cell activation after phagocytosis of necrotic trophoblastic material: implications for the pathogenesis of pre-eclampsia.

Qi Chen; Peter Stone; Lai-Ming Ching; Lawrence W. Chamley

Pre‐eclampsia is characterized by systemic maternal endothelial dysfunction that precedes the onset of clinical symptoms. The cause of the dysfunction is not clear but the number and the nature of trophoblasts shed from the placenta may be altered in pre‐eclamptic pregnancies. These dead trophoblasts become trapped in the pulmonary capillaries and may then be phagocytosed by endothelial cells. Phagocytosis of necrotic, but not apoptotic, trophoblasts results in endothelial cell activation. We have explored the hypothesis that activation can subsequently spread to other endothelial cells via soluble factors without the need for direct contact with shed trophoblasts. Conditioned medium from endothelial cells that had phagocytosed necrotic, but not apoptotic, trophoblasts was shown to activate fresh endothelial cells due, in large part, to IL‐6 secreted into the conditioned medium. The amount of IL‐6 secreted in response to phagocytosis of necrotic trophoblasts was similar to the levels of IL‐6 found by others in the blood of pre‐eclamptic women and was substantially more than the level of IL‐6 which has been reported to induce symptoms of pre‐eclampsia in pregnant rats. We demonstrated that phagocytosis of both a trophoblast cell line as well as trophoblasts shed from human placentae, had this effect on two different types of endothelial cells. The role of IL‐6 in endothelial cell activation was confirmed using recombinant IL‐6 and neutralizing antibodies against IL‐6 and the IL‐6 receptor. Thus, IL‐6 secreted by pulmonary endothelial cells after they have phagocytosed necrotic trophoblasts that are trapped in the pulmonary capillaries could activate endothelial cells in other remote vascular beds, contributing to the systemic activation of the endothelium that is a hallmark of pre‐eclampsia. Copyright

Collaboration


Dive into the Lai-Ming Ching'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

Petr Tomek

University of Auckland

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge