34
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

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

      research-article

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          5,6-dimethylxanthenone-4-acetic acid (DMXAA), a new anticancer agent synthesised in this laboratory (Rewcastle et al, 1991), is currently undergoing clinical evaluation as an antivascular agent for the treatment of cancer. In mice with transplantable tumours, DMXAA caused cessation of tumour blood flow, vascular collapse and tumour necrosis (Rewcastle et al, 1991; Zwi et al, 1994; Lash et al, 1998). DMXAA also increased tumour necrosis factor (TNF) concentrations in both plasma and tumour tissue of mice (Philpott et al, 1995; Ching et al, 1999). We have previously used TdT-mediated dUTP nick-end labelling (TUNEL) assays to demonstrate the induction of apoptosis of the vascular endothelium in Colon 38 tumours in mice treated with DMXAA at its optimal dose (Ching et al, 2002). Staining was detectable within 30 min of administration, intensified with time, and necrosis of adjacent tumour tissue was evident after 3 h. Some apoptosis of splenic tissue was detected in tumour-bearing mice, but none was observed in the liver tissue. Of particular interest was the finding of TUNEL staining of tumour vascular endothelium in breast tumour biopsies taken from a patient 3 and 24 h after infusion of DMXAA (3100 mg m−2) in a Phase I clinical trial. Thus, DMXAA is capable of inducing apoptosis in vascular endothelial cells in both mice and human tumours. The finding of a rapid onset of tumour endothelial apoptosis, occurring before the appearance of detectable TNF in tumour tissue (Ching et al, 1999), suggests that DMXAA exerts a direct effect on tumour vasculature, and is of particular relevance to clinical trials. In this report, we have used in vivo vascular labelling techniques to investigate the relationship between apoptosis induction and tumour blood flow reduction. To investigate the role of TNF, we utilised mice with a targeted disruption of the TNF gene (TNF−/−) or of the TNF receptor 1 gene (TNFR−/−). MATERIALS AND METHODS Materials DMXAA was synthesised at the Auckland Cancer Society Research Centre (Rewcastle et al, 1991) and dissolved in minimal 5% sodium bicarbonate for intraperitoneal injection into mice (25 mg kg−1) in a volume of 0.01 ml g−1 body weight. Hoechst 33342 (Sigma Chemical Co., St Louis, MO, USA) was dissolved at 8 mg ml−1 in saline and stored at −80°C. Mice All mice were housed and used under institutional, ethical guidelines. All animal experiments have been carried out with ethical committee approval. The ethical guidelines that were followed meet the standards required by the UKCCCR guidelines (Workman et al, 1998). C57Bl/6 mice were obtained from the Animal Resource Unit, University of Auckland. TNF−/− and TNFR−/− knockout mice on a C57Bl/6 background were offspring from breeding pairs obtained, respectively, from the Centenary Institute, Sydney, Australia, and Jackson Laboratory, Bar Harbor, ME, USA. Colon 38 tumour fragments (1 mm3) were implanted subcutaneously in the left flank of anaesthetised (82 mg kg−1 sodium pentobarbitone) mice. Tumours were used when they had reached approximately 6 mm in diameter, generally 10 days after implantation. At least three mice were assigned for each group. Histochemistry Apoptosis was determined using the TUNEL assay for the identification of double-stranded DNA breaks using the In situ Cell Death Detection Kit (Roche Diagnostics, Mannheim, Germany), according to the manufacturer's instructions. Tissue cryosections (14 μm thickness) on poly-L-lysine-coated slides were fixed in 4% paraformaldehyde in phosphate-buffered saline (PBS) for 30 min at room temperature, washed three times with PBS for 10 min each time, dehydrated for 2 min in absolute ethanol and then treated with permeabilisation solution (1% Triton X-100 in 1% sodium citrate) for 15 min at room temperature. Strand breaks were labelled with fluoresceinated dUTP and visualised following reaction with either antifluorescein antibody conjugated with alkaline phosphatase and Vector® Black alkaline phosphatase substrate solution (Vector Laboratories, Burlingame, CA, USA) or antifluorescein antibody conjugated with horseradish peroxidase (POD) and diaminobenzidine (DAB) substrate (Roche Diagnostics, Mannheim, Germany). All slides were counter stained using methyl green. The amount of apoptotic staining in the sections was quantitated using Adobe Photoshop, Version 4 (Adobe Systems Inc., San Jose, CA, USA). For each of 5–10 random fields of tumour sections (2–3 tumours per group), the number of pixels stained with TUNEL was determined, divided by the total number of pixels, and expressed as a percentage. Tissue cryosections were also fixed in cold acetone for 20 min at 4°C, blocked with 1.5% normal rabbit serum for 1 h at room temperature, incubated with avidin–biotin for 15 min, and then incubated with 1 : 100 dilution of rat anti-mouse CD-31 monoclonal antibody (MEC 13.3; BD Pharmingen, USA) overnight at 4°C in a humidified container. Sections were then incubated with 1 : 100 dilution of biotinylated anti-rat IgG antibody and avidin–biotin complex (Vectastain ABC-AP Kit, Vector Laboratories, Burlingame, CA, USA). Immunoglobulin complexes were visualised using Vector Red alkaline phosphatase substrate solution, also from Vector Laboratories. Hoechst 33342 staining of functional vessels Hoechst 33342 (8 mg ml−1 in saline) was injected via the tail vein at 0.1 ml per mouse 3 h after DMXAA treatment. Mice were killed 2 min later by cervical dislocation and the tumours were excised and frozen at −80°C. Cryosections (14 μm) of the tumour were examined using a fluorescence microscope with a UV-1A filter block (excitation 365 nm, barrier filter 400 nm, dichroic mirror 400 nm). Five–10 fields per tumour were scored (two to three tumours per group), and the number of positively stained vessels per 1 mm−2 field was calculated. Statistical analyses Data were analysed using a paired Student's t-test and by standard correlation analysis. A probability value of <0.05 was considered significant. RESULTS Endothelial cell apoptosis in various tissues following DMXAA Sections of Colon 38 tumours, liver, spleen, heart and brain collected from C57Bl/6 mice without treatment or 3 h after DMXAA administration (25 mg kg−1) were stained for apoptosis using TUNEL (Figure 1 Figure 1 Selective induction of tumour vascular endothelial cell apoptosis by DMXAA. Sections from Colon 38 tumours, spleen, liver, heart and brain from untreated or treated (DMXAA, 25 mg kg−1, 3 h) C57Bl/6 mice were stained for TUNEL with alkaline phosphatase substrate (A–J) or POD/DAB (K–T). Stained sections shown at × 100 magnification. ). Tumour sections from DMXAA-treated mice showed 12.5- and 12-fold increases in apoptosis staining over that in tumour sections from untreated mice using alkaline phosphatase, or POD/DAB, respectively, as the enzyme system for visualisation of apoptosis staining (Figure 2 Figure 2 Apoptosis after 3 h in Colon 38 tumours, spleen, liver, heart and brain from mice treated with 25 mg kg−1 DMXAA. Bars represent ratios of percentage TUNEL-stained areas in the treated tissue to that in untreated tissue. Alkaline phosphatase substrate (black bars); POD/DAB substrate (grey bars). ). No statistically significant increases in apoptosis staining were observed in other tissues (Figure 2). Liver sections from treated or untreated mice showed no staining. Using alkaline phosphatase, false-positive background staining was observed in the spleen, heart and brain sections of tumour-bearing and nontumour-bearing mice, and in negative control sections that had not been incubated with the immunohistochemistry reagents. Staining of sections of brain, liver, heart or spleen from treated or untreated mouse was not observed using the DAB substrate system, which confirmed that induction of apoptosis following DMXAA treatment was specific to tumour tissue (Figure 1). Similar ratios of apoptosis induction in tumour tissues were obtained using either POD/DAB or alkaline phosphatase (Figure 2), but the latter produced more intense labelling and was used in subsequent studies with tumour tissues. To ascertain whether the apoptotic cells were endothelial cells, adjacent cryosections were stained with antibodies to CD-31 and the pattern of staining with the anti-CD-31 and apoptosis compared. Similar staining patterns were observed, providing strong evidence for DMXAA-induced endothelial apoptosis. Dose–response relationship and time course of DMXAA-induced tumour endothelial cell apoptosis and blood flow inhibition A significant increase in apoptotic vessels in Colon 38 tumour sections, analysed 3 h after DMXAA treatment, was seen at doses as low as 5 mg kg−1 (Table 1 Table 1 Apoptosis induction and blood flow inhibition in Colon 38 tumours following DMXAA treatment Treatment   Endothelial cell apoptosis Blood flow inhibition DMXAA (mg kg−1) Time (h) TUNEL stain (% area) Increase over untreated Hoechst-stained vessels mm−2 Percentage inhibition 0 0 0.2±0.1   29.9±1.4   5 3 0.5±0.1 2.5 (0.003) 28.8±1.2 4 (0.6) 10 3 0.8±0.3 4.0 (0.005) 26.2±1.3 13 (0.9) 15 3 1.0±0.2 5.0 (<0.001) 13.4±1.0 56 (<0.001) 20 3 1.2±0.1 6.0 (<0.001) 10.2±1.0 66 (<0.001) 25 3 2.5±0.9 12.5 (0.003) 7.2±0.7 76 (<0.001) 25 1 1.2±0.1 6.0 (<0.001) 12.5±0.6 58 (<0.001) 25 0.5 0.6±0.2 3.0 (<0.001) 18.4±1.2 39 (<0.001) 25 0.25 0.5±0.2 2.5 (0.003) 25.8±1.5 14 (0.08) DMXAA=5,6-dimethylxanthenone-4-acetic acid. P-values in brackets represent the degree of statistical difference between treated and untreated controls. , Figure 3A Figure 3 Apoptosis and blood flow measured in Colon 38 tumours treatment with DMXAA at different doses after 3 h (A), or at different times after DMXAA at a dose of 25 mg kg−1 (B). Percentage TUNEL-stained areas (Δ); Hoechst-stained vessels mm−2 (▴). ). The frequency of apoptotic vessels increased with increasing dose, with a particularly sharp increase from 20 mg kg−1 (six-fold induction as compared to untreated controls) to the MTD of 25 mg kg−1 (12.5-fold induction; Table 1). Apoptosis of tumour vascular endothelial cells was detectable as early as 15 min (2.5-fold increase) and progressively increased with time following administration of DMXAA at the MTD (Table 1, Figure 3B). As a measure of blood flow inhibition following DMXAA treatment, we used the perfusion marker Hoechst 33342 to stain functional vessels (Zwi et al, 1989). No inhibition of blood flow was observed after 3 h with DMXAA doses of 5 and 10 mg kg−1. Inhibition was 56% at a dose of 15 mg kg−1 and increased progressively with dose up to the MTD (Table 1, Figure 3A). Blood flow was significantly reduced (39%) 30 min after DMXAA treatment at 25 mg kg−1, and reached 76% inhibition after 3 h (Table 1, Figure 3B). DMXAA-induced tumour endothelial cell apoptosis and blood flow shutdown in TNF−/− and TNFR−/− mice To determine if the antivascular effects of DMXAA were TNF-dependent, we compared the responses in TNF−/− and TNFR−/− mice to those in wild-type C57Bl/6 mice. Tumour endothelial cell apoptosis in TNF−/− and TNFR−/− hosts following DMXAA (25 mg kg−1) was, respectively, 1.8- and 10.4-fold lower than that in wild-type mice. However, the knockout mice tolerated higher doses of DMXAA and, at a dose of 50 mg kg−1, the induced apoptosis was comparable to that obtained in wild-type mice at 25 mg kg−1 of DMXAA in wild-type mice. Blood flow in tumours implanted in TNF−/− and TNFR−/− mice was determined from Hoechst-stained vessels, and was, respectively, 2.5- and 5.3-fold lower than that in tumours in wild-type mice, 3 h following DMXAA at 25 mg kg−1. Again, however, at the higher dose of 50 mg kg−1, which can be tolerated by the knockout mice, inhibition of blood flow was similar to that obtained at 25 mg kg−1 in wild-type mice (Table 2 Table 2 Endothelial cell apoptosis and blood flow inhibition in tumours from C57Bl/6, TNF–/− and TNFR–/− mice following DMXAA treatment   Percentage TUNEL-stained areas Hoechst-stained vessels mm−2 DMXAA C57Bl/6 TNF−/− TNFR−/− C57Bl/6 TNF−/− TNFR−/− 0 0.2±0.1 0.2±0.1 0.3±0.1 29.9±1.4 32.5±1.7 35.7±2.7 25 mg kg−1 2.5±1.0 1.4±0.5 0.3±0.1 5.6±1.0 13.6±1.4 31.3±1.3 50 mg kg−1 — 2.8±0.8 1.9±0.6 — 6.0±0.5 6.0±0.6 DMXAA=5,6-dimethylxanthenone-4-acetic acid. ). DISCUSSION These results confirm our previous findings (Ching et al, 2002) that DMXAA induces endothelial cell apoptosis in Colon 38 tumours. Apoptosis induction was selective to tumour vascular endothelium and was not seen in liver, heart, brain or spleen (Figures 1 and 2). We had previously reported apoptosis staining in splenic tissues, using alkaline phosphatase for the detection of the bound antibodies (Ching et al, 2002), but the results here show that the staining observed in the normal organs using the alkaline phosphatase procedure was not DMXAA-induced and was likely to be due to high endogenous phosphatase levels that had not been completely blocked (Figure 2). The basis for the pronounced selectivity for tumour vasculature is not yet understood. Factors secreted by tumour-associated immune cells, or by the tumour cells themselves, may play a role by ‘priming’ the response of tumour endothelial cells to DMXAA. Tumour-conditioned medium has been reported to play a role in modulating the response of cultured endothelial cells to flavone acetic acid (Watts and Woodcock, 1992). Endothelial cells in culture are resistant to apoptosis induction by DMXAA (Ching et al, 2002), and we have found that addition of serum from Colon 38-bearing mice did not render them sensitive (unpublished results). To determine whether there was a relationship between the degree of blood flow inhibition and endothelial cell apoptosis induction, all the data for both wild-type and knockout mice treated with DMXAA with different doses and at different times were plotted on the same graph (Figure 4 Figure 4 Relationship between the logarithm of induced apoptosis and the logarithm of blood flow inhibition in Colon 38 tumours, plotted for all the experiments. Wild-type C57Bl/6 (○); TNF−/− (Δ); TNFR−/− (□). ). A highly significant logarithmic relationship was found (r=0.94; P<0.001), indicating that a 10% increase in apoptosis leads to a 7% decrease in blood flow. The degree of significance suggests that tumour blood flow inhibition is a consequence of endothelial cell apoptosis. Damage to the endothelium and subsequent loss of the structural integrity of the vessels leading to increase in vascular permeability would result in a reduction in blood flow (Baguley, 2003). TNF is induced following DMXAA administration to mice (Philpott et al, 1995), and the histology of tumours treated with DMXAA resembles that of TNF-treated tumours, suggesting that TNF participates in the antivascular action. Support for this hypothesis is provided by experiments where Colon 38 tumours were implanted in TNF−/− and TNFR−/− knockout mice, where the antitumour effects following administration of the same dose of DMXAA are substantially reduced (Ching et al, 1999; Zhao et al, 2002). In agreement with these findings, apoptosis induction and tumour blood flow inhibition following treatment with DMXAA (25 mg kg−1) were pronounced in tumours implanted in wild-type mice, but small in tumours implanted in TNF−/− and TNFR−/− knockout mice (Table 2). The lower toxicity of DMXAA in these knockout mice allows the use of higher drug doses, which restored both apoptosis induction and tumour blood flow inhibition responses. The results are consistent with the hypothesis that DMXAA can exert an antivascular response both directly and indirectly by induction of TNF, and perhaps of other cytokines. The relationship in Figure 4 suggests that both direct and indirect mechanisms act with a similar relationship between apoptosis induction and tumour blood flow inhibition. These results are of particular importance to clinical studies, since TNF levels were not found to be raised in Phase I clinical trials of DMXAA but tumour blood flow shutdown at doses above 500 mg m−2 was clearly demonstrable (Rustin et al, 1998; Jameson et al, 2003). Multiple mediators of antivascular effects may be involved in providing a selective antitumour effect.

          Related collections

          Most cited references14

          • Record: found
          • Abstract: found
          • Article: not found

          Antivascular therapy of cancer: DMXAA.

          The vascular endothelium of tumour tissue, which differs in several ways from that of normal tissues, is a potential target for selective anticancer therapy. By contrast with antiangiogenic agents, antivascular agents target the endothelial cells of existing tumour blood vessels, causing distortion or damage and consequently decreasing tumour blood flow. DMXAA (5,6-dimethylxanthenone-4-acetic acid), a low-molecular-weight drug, has a striking antivascular and in some cases curative effect in experimental tumours. Its action on vascular endothelial cells seems to involve a cascade of events leading to induction of tumour haemorrhagic necrosis. These events include both direct and indirect effects, the latter involving the release of further vasoactive agents, such as serotonin, tumour necrosis factor, other cytokines, and nitric oxide from host cells. Phase I clinical trials of DMXAA have been completed and the next challenge to face is how the antivascular effect of this drug should be exploited for the treatment of human cancer.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

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

            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. British Journal of Cancer (2002) 86, 1937–1942. doi:10.1038/sj.bjc.6600368 www.bjcancer.com © 2002 Cancer Research UK
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Clinical aspects of a phase I trial of 5,6-dimethylxanthenone-4-acetic acid (DMXAA), a novel antivascular agent

              Targeting tumour vasculature as a treatment for cancer is the subject of much research, with most effort currently directed at angiogenesis inhibitors. Yet therapies that interrupt existing tumour vasculature and result in haemorrhagic necrosis of tumours have been investigated for over a century and were mostly associated with bacterial infections or their toxins (Chaplin and Dougherty, 1999). William Coley, who pursued the method avidly from 1891 (Council on Pharmacy and Chemistry, 1934), achieved significant cure rates, especially in sarcoma and lymphoma, using various bacterial toxins both for advanced disease and as adjuvant therapy (Coley-Nauts et al, 1953). Coley's toxins were abandoned in favour of radiotherapy, but more recent clinical studies with bacterial endotoxins have shown modest antitumour activity, although systemic toxicity remains dose-limiting (Otto et al, 1996; DeVore et al, 1997). In 1947, investigators at the National Cancer Institute observed that a toxin from the bacterium Serratia marcescens acutely and irreversibly reduced blood flow to sarcomas in mice, while blood flow to muscle recovered within 18 h (Algire et al, 1947). The induction of tumour necrosis factor (TNF) by bacterial toxins was later shown to be the cause of haemorrhagic necrosis in tumours (Carswell et al, 1975; Old, 1985). Clinical trials of recombinant human TNF were unable to deliver therapeutic doses systemically because of toxicity (Spriggs and Yates, 1992), but TNF is highly effective in conjunction with chemotherapy in isolated limb perfusion for melanoma and sarcoma, where it has been shown to have selective tumour vascular effects (Lejeune et al, 1998; Ruegg et al, 1998; Eggermont and ten Hagen, 2001). Nonbacterial, tumour vascular-targeting agents could have advantages over bacterial products in terms of toxicity and pharmacology. One such agent, flavone acetic acid (FAA), showed remarkable anticancer activity in murine models (O'Dwyer et al, 1987) but its activity was minimal in clinical trials, although these were conducted when it was thought to be directly cytotoxic and its indirect mechanisms of antitumour action were poorly understood (Kerr and Kaye, 1989; Bibby, 1991). Further preclinical studies at the Auckland Cancer Society Research Centre (ACSRC) showed that FAA induced TNF production, acute tumour vascular collapse, haemorrhagic necrosis of tumours and enhanced activity of immune effector cells (Ching and Baguley, 1987; Smith et al, 1987; Finlay et al, 1988; Zwi et al, 1989, 1992) (Zwi et al, 1990 ID: 286). Few appropriate pharmacodynamic (PD) studies were performed in the clinical trials of FAA, so it is not known whether it has significant antivascular or immunological activity in humans. However, in vitro studies suggested a species difference in antitumour activity, because FAA induced TNF in murine, but not human, peripheral blood mononuclear cells (Ching et al, 1994; Philpott et al, 1997). 5,6-Dimethylxanthenone-4-acetic acid (DMXAA), a new analogue of FAA developed in the ACSRC, showed greater activity and 12-fold higher dose-potency than FAA in murine tumour models (Rewcastle et al, 1991) and appeared to overcome the species difference in in vitro TNF production (Ching et al, 1994; Philpott et al, 1997). Dose-limiting toxicity in mice was consistent with hypotension, and was not strictly related to TNF production (Philpott et al, 1995). While DMXAA proceeded into clinical development because of this favourable profile, further studies in animal tumour models have demonstrated synergistic interactions between DMXAA with radiotherapy (Wilson et al, 1998), chemotherapy (particularly taxanes) (Pruijn et al, 1997; Horsman et al, 1999; Wilson and Baguley 2000; Siim et al, 2002), bioreductive cytotoxic drugs (Cliffe et al, 1994; Lash et al, 1998), radioimmunotherapy (Pedley et al, 1996), antibody-directed enzyme prodrug therapy (ADEPT) (Pedley et al, 1999), thalidomide (Cao et al, 1999) and immunotherapy (Kanwar et al, 2001). Cancer Research UK selected DMXAA for two parallel phase I trials with different schedules, one in the UK using a weekly administration schedule (at Mt Vernon Hospital, Northwood, and Bradford Royal Infirmary, Bradford) and the other in Auckland, NZ, dosing every 3 weeks. The objectives of the NZ study were to determine the toxicity of DMXAA, its maximum tolerated dose (MTD), pharmacokinetics (PK), selected PD end points and, as a secondary objective, antitumour efficacy. A protocol amendment permitted evaluation of changes in tumour blood flow by dynamic magnetic resonance imaging (MRI) and the findings (including both UK and NZ trial patients) have recently been published (Galbraith et al, 2002). This report focuses on clinical aspects of the NZ trial; PD and PK aspects, due to their complexity, will be reported separately. MATERIALS AND METHODS Patients Eligibility criteria included: patients ⩾18 years with histologically or cytologically proven cancer refractory or not amenable to conventional therapy; WHO performance status of 0–2; life expectancy >3 months; haemoglobin ⩾9 g dl−1, WBC ⩾3 × 109 l−1, platelets ⩾100 × 109 l−1; creatinine ⩽130 μmol l−1; bilirubin within normal limits, ALT and alkaline phosphatase 450 ms in males, >470 ms in females) after treatment with doses of DMXAA of ⩾3100 mg m−2 (Committee for Proprietary Medicinal Products, 1997). The maximal QTc prolongation occurred during or within 15 min of completion of the drug infusion with a return to baseline generally over 4–6 h (Figure 2 Figure 2 Change in corrected QT interval after DMXAA administration at 3700 mg m−2. ). QTc was prolonged beyond 500 ms in four patients with baseline values of 430, 460, 461 and 501 ms and in the first three patients, this lasted for less than 1 h. No clear dose–response relationship was observed and no ventricular tachyarrhythmia was seen. Dyspnoea possibly related to DMXAA was documented in three patients treated at doses ⩽1100 mg m−2, but each patient had another more likely explanation. At higher doses, acute dyspnoea at rest occurred in four patients immediately following DMXAA infusion. In three of these patients (treated at 2600–3700 mg m−2), the dyspnoea after DMXAA was minimal, brief (5–25 min) and clinically insignificant. The fourth patient, with moderately severe chronic obstructive respiratory disease, became very breathless and anxious 15 min after her first course at 4900 mg m−2. Clinically, she had basal pulmonary inspiratory crackles but no bronchospasm. Oxygen saturation was normal on breathing air and the symptoms resolved in about 75 min. On her second course, DMXAA was reduced to 3700 mg m−2 (she is thus represented at two doses in Table 3) and she again developed dyspnoea, lasting for about an hour. She had basal inspiratory crackles and a third heart sound (consistent with left ventricular failure) and these signs resolved within 2 h. Therapeutic response A total of 60 patients were evaluable for response. One partial response was seen in a patient with metastatic squamous carcinoma of cervix treated at 1100 mg m−2 and previously treated with bleomycin, etoposide and cisplatin chemotherapy, then carboplatin. The response was unconfirmed because two small neck nodes increased in size transiently after the third course then subsequently regressed again and overall tumour response was maintained for eight courses (Figure 3 Figure 3 Tumour response in a patient with metastatic squamous carcinoma of cervix treated with DMXAA 1100 mg m−2. Tumour size was calculated as the sum of the products of bidimensional measurements of three clinically measurable metastatic neck nodes. ). No change was the best response in 22 patients (35%) at dose levels from 6 to 3700 mg m−2 and the duration exceeded 12 weeks in five patients (8%). DISCUSSION This phase I trial of DMXAA has demonstrated clinical antitumour activity and reduction in tumour blood flow at well-tolerated doses (Galbraith et al, 2002). The MTD was established as 3700 mg m−2, and higher doses produced unusual, transient toxicities including confusion, slurred speech, tremor, restlessness and visual disturbance, as well as transient autonomic changes such as sweating, tremor, changes in blood pressure and heart rate. The results of the UK trial of DMXAA are published separately (Rustin et al, submitted to Br J Cancer), but there was a high degree of concordance with our results including toxicities, PK, PD and clinical activity (an unconfirmed partial response was also seen). Clinical trials of FAA, which is structurally related to DMXAA, showed differences between the two drugs. While the clinical toxicities of DMXAA and FAA showed some similarities (warmth, flushing, sweating, fatigue, myalgia, nausea, vomiting and visual disturbance), the DLTs of FAA were remarkably different and included hypotension, diarrhoea, flushing, asthenia and fatigue (Kerr et al, 1987; Weiss et al, 1988; Kaye et al, 1990; Havlin et al, 1991; Olver et al, 1992). Prolongation of bleeding time was described (Rubin et al, 1987) and one patient presented with haemorrhage due to immune thrombocytopenia after five doses of FAA (Davis et al, 1988). Bleeding time was not measured in this DMXAA trial, but no significant disturbance of INR and APTT was seen, nor was any significant thrombocytopenia observed. A number of other agents, including serotonin, some tubulin-binding agents and arsenic trioxide, selectively inhibit tumour blood flow (Stücker et al, 1992; Ching et al, 1999; Lew et al, 1999). Combretastatin A-4 phosphate (CA4P), a tubulin-binding agent, has antivascular activity at doses well below the MTD in both preclinical and early clinical studies (Rustin et al, 1999), comparable to those reported with DMXAA (Lash et al, 1998; Galbraith et al, 2002). However, the blood flow-modifying effect of CA4P is not entirely tumour-selective (Griggs et al, 2001), and it appears to be reversible except at high doses (Anderson et al, 2000; Murata et al, 2001). Some CA4P toxicities resembled those of DMXAA (including flushing, nausea, vomiting, tumour pain and QTc prolongation), but others (cardiac ischaemia and cerebellar ataxia) were notably different (Galbraith et al, 2001; Dowlati et al, 2002). Some toxicities observed with DMXAA resemble the ‘serotonin syndrome’, attributed to high levels of serotonin in the central nervous system (CNS). This syndrome most commonly occurs when two drugs are taken, which can each increase CNS serotonin and includes alterations in cognition, behaviour, autonomic nervous system function and neuromuscular activity (Sporer, 1995). Supportive evidence comes from two other aspects of this trial: firstly, a patient took a monoamine oxidase inhibitor prior to treatment with DMXAA at 1650 mg m−2 and developed clinical features of the serotonin syndrome; and secondly an acute increase in plasma prolactin levels seen in many patients treated with DMXAA at ⩾2000 mg m−2 (unpublished results). This occurs following production of serotonin in the CNS (Van de Kar et al, 1996). If subsequent studies provide further evidence that DMXAA increases CNS serotonin release, it would be prudent to avoid administering DMXAA to patients who are receiving other drugs that increase CNS serotonin levels (Brown et al, 1996). The mechanism underlying the acute release of CNS serotoninis not known, but serotonin release in plasma has been observed in this study (Kestell et al, 2001), and is a feature of the antivascular activity of this drug in preclinical models (Baguley et al, 1997). The visual toxicities of DMXAA are not a feature of the serotonin syndrome, but the blurring, colour disturbance and photophobia have similarities to those reported with sildenafil (Viagra®, Pfizer, New York, NY, USA) (Marmor and Kessler, 1999). The latter's visual toxicities are thought to be due to inhibition of phosphodiesterase type 6 (PDE6), which exists exclusively in the retina and is responsible for modulating the transduction cascade of the photoreceptor response to light. Recent data have shown that DMXAA inhibited PDE6 in vitro at pharmacologically relevant concentrations (e.g. 50 μ M) (personal communication, L Kelland, Antisoma plc). It is reassuring in this regard that no long-term retinal sequela of sildenafil administration is known, including data from retinal histologic studies in dogs dosed with 65 times the maximum recommended human dose daily for 12 months (Wallis et al, 1998; Marmor and Kessler, 1999). The significance of the observed QTc prolongation in these preliminary data is uncertain given its brevity, its possible relation to autonomic changes caused by the infusion and the population of patients potentially being treated with this agent. Moreover, the use of ad hoc heart rate correction formulae (such as Bazett's) may bias the result (Aytemir et al, 1999). Therefore the QTc results in this trial must be regarded as indicative only. However, given the concern that QT interval prolongation may predispose to ventricular tachycardia (Committee for Proprietary Medicinal Products, 1997), it will be important to determine the dose–response relationship of QTc prolongation before deciding on the dose of DMXAA for phase II and combination studies. In conclusion, DMXAA is well tolerated over a wide dose range and has clinical and biological features distinct from those of other antivascular drugs in clinical development. Further clinical trials with this agent are clearly warranted, particularly in combination with other treatment modalities where synergistic interactions are observed in animal tumour models.
                Bookmark

                Author and article information

                Journal
                Br J Cancer
                British Journal of Cancer
                Nature Publishing Group
                0007-0920
                1532-1827
                17 February 2004
                23 February 2004
                : 90
                : 4
                : 906-910
                Affiliations
                [1 ] 1Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland, New Zealand
                Author notes
                [* ]Author for correspondence: l.ching@ 123456auckland.ac.nz
                Article
                6601606
                10.1038/sj.bjc.6601606
                2410181
                14970872
                6fcd0414-553c-475e-9ef8-0d5fadfc459b
                Copyright 2004, Cancer Research UK
                History
                : 22 September 2003
                : 27 October 2003
                Categories
                Experimental Therapeutics

                Oncology & Radiotherapy
                antivascular,apoptosis,knockout mice,blood flow,tumour necrosis factor,hoechst 33342

                Comments

                Comment on this article