Skip to main content
  • AACR Journals
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Radiation Oncology
      • Novel Combinations
      • Reviews
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Journals
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • My Cart

Search

  • Advanced search
Molecular Cancer Therapeutics
Molecular Cancer Therapeutics
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Radiation Oncology
      • Novel Combinations
      • Reviews
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Article

Modulation of chemotherapy resistance in regional therapy: a novel therapeutic approach to advanced extremity melanoma using intra-arterial temozolomide in combination with systemic O6-benzylguanine

Tomio Ueno, Sae Hee Ko, Elizabeth Grubbs, Yasunori Yoshimoto, Christi Augustine, Zeinab Abdel-Wahab, Tsung-Yen Cheng, Omar I. Abdel-Wahab, Scott K. Pruitt, Henry S. Friedman and Douglas S. Tyler
Tomio Ueno
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Sae Hee Ko
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Elizabeth Grubbs
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Yasunori Yoshimoto
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Christi Augustine
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Zeinab Abdel-Wahab
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Tsung-Yen Cheng
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Omar I. Abdel-Wahab
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Scott K. Pruitt
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Henry S. Friedman
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Douglas S. Tyler
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1158/1535-7163.MCT-05-0098 Published March 2006
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

This study investigated whether the therapeutic index of regional melanoma therapy using parenteral temozolomide could be improved by chemomodulation with O6-benzylguanine (O6BG), an inhibitor of the DNA repair enzyme O6-alkylguanine-DNA alkyltransferase (AGT). Using a nude rat s.c. human melanoma xenograft model of the extremity, tumors were analyzed for AGT level 2 to 3 hours after the i.p. injection of 3.5 to 70.0 mg/kg O6BG to inhibit AGT activity. Survival studies were conducted using animals that were treated with a 15-minute isolated limb infusion with 10% DMSO in PBS (control), temozolomide alone, or temozolomide in conjunction with single or multiple doses of i.p. O6BG. Tumor volume and toxicity level were monitored every other day. Administration of 3.5 mg/kg O6BG depleted tumor AGT activity by 93.5% (P < 0.01). Groups treated with regional temozolomide alone (350 mg/kg), systemic temozolomide with O6BG, or vehicle combined with O6BG showed no significant tumor responses compared with controls. Whereas use of regional temozolomide alone at a higher dose (750 mg/kg) showed some degree of tumor response, regional temozolomide given in conjunction with multiple dosages of O6BG showed a marked (P < 0.01) reduction in tumor growth with minimal toxicity. Our findings suggest that AGT modulation by the administration of O6BG in combination with temozolomide regional chemotherapy leads to a significant improvement in melanoma antitumor responses. Clinical trials using chemotherapy modulation may improve response rates in future regional infusion and perfusion drug trials. [Mol Cancer Ther 2006;5(3):732–8]

Keywords:
  • Regional therapy
  • Melanoma
  • Temozolomide
  • O6-alkylguanine-DNA alkyltransferase (AGT)
  • O6-benzylguanine (O6BG)

Introduction

Regional failure in patients with extremity melanoma is a significant problem. Aggressive therapy in the form of amputation suggests that 25% to 35% of these patients may have disease confined to the extremity (1). Isolated limb infusion or perfusion with cytotoxic agents is frequently used for this clinical problem. Melphalan (l-phenylalanine mustard) has been accepted as the most widely used drug in the regional setting for advanced extremity melanoma although many cytotoxic drugs have been studied. Recently, however, we have shown in a clinically relevant animal model of regionally advanced melanoma that regional temozolomide has better antitumor activity than either systemic temozolomide or regional melphalan (2).

Attempts at modulation of mechanisms that interfere with the optimal effectiveness of chemotherapeutic agents have been explored previously in vitro, but in vivo experiments have been limited due to increased systemic toxicity when chemotherapy and modulators are used together (3). Utilization of these strategies in a regional setting has the advantage of minimizing the systemic side effects of combination therapy while maximizing the effect regionally on the tumor. We have previously investigated modulation of putative resistance mechanisms in the context of regional therapy for melanoma and showed marked improvements in the therapeutic index of melphalan by interfering with the glutathione detoxification system (3–5). The specific aim for this project was to define if the therapeutic index of regional melanoma therapy with temozolomide can be enhanced using systemic inhibition of O6-alkylguanine-DNA alkyltransferase (AGT), the predominant mechanism of resistance to this methylating agent (6), by administration of the drug O6-benzylguanine (O6BG) in an animal model that parallels the clinical situation of in-transit disease in humans.

Materials and Methods

Animals and Tumor Inoculation

Female athymic nude rats (age, 5–7 weeks; RHU-M, Harlan, Indianapolis, IN) were housed in a temperature-controlled room with a 12-hour light-dark cycle. A standard laboratory diet and water were provided ad libitum.

Within 3 days after irradiation at a dose of 500 cGy to facilitate growth of human melanoma xenografts, the rats were injected s.c. in the right distal hind limb just proximal to the ankle with 5 × 106 culture human melanoma cells (DM6; ref. 3). Xenografts were measured every other day with vernier calipers in two perpendicular dimensions and tumor volume was calculated according to the following formula: [(length) × (width)2] / 2 (5). All aspects of the experimental protocol were approved by the Durham Veterans Affairs Medical Center Institutional Animal Care and Use Committee.

O6BG Dosing Study

We routinely prepared fresh O6BG stock solution of 2 mg/mL in 40% polyethylene glycol-400 (PEG) in PBS. The O6BG solution of interest was then made by further dilution of O6BG stock solution with 40% PEG in PBS according to the given dosing regimen. A solution of 40% PEG in PBS was used as the vehicle control for the O6BG experiments.

To evaluate the inhibitory effect of O6-benzylguanine (O6BG; Sigma Chemical Co, St. Louis, MO) on AGT activity in the regional nude rat human melanoma xenograft model, we compared tumor and muscle AGT levels from four groups of rats with a similar size xenograft by giving 3.5, 17.5, 35, or 70 mg/kg O6BG i.p. (n = 3 in each group). Two to three hours after the administration of O6BG, both tumor and muscle tissues were harvested, immediately snap frozen, and analyzed for AGT. Samples were removed under general anesthesia to prevent the loss of enzyme activity. To measure the baseline AGT activity of tumor and muscle, tissues were harvested from four rats 2 to 3 hours after i.p. administration vehicle (40% PEG in PBS).

AGT Determination

Samples of harvested tumor xenografts were weighed and extracts were prepared by adding 2 mL of 50 mmol/L Tris (pH 7.5), 0.1 mmol/L EDTA, and 5 mmol/L DTT buffer per gram of tissue weight. Samples were homogenized for 1 minute, sonicated for 1 minute, and centrifuged at 14,000 × g for 30 minutes. The assay for AGT activity was done as previously described by Domoradski et al. (7). Briefly, AGT activity was measured as the removal of O6-[3H]-methylguanine from a [3H]-methylated DNA substrate (5.8 Ci/mmol) following incubation with tissue extract at 37°C for 30 minutes. The DNA was precipitated by adding ice-cold perchloric acid (0.25 N) and hydrolyzed by the addition of 0.1 N HCl at 70°C for 30 minutes. Following filtration using a microfilter apparatus, the modified bases are separated by reverse-phase high-performance liquid chromatography with 0.5 mol/L ammonium formate (pH 4.5) containing 5.5% methanol. Protein is determined by the method of Bradford and the amount of O6-methylguanine released from the DNA substrate per mg of protein was calculated. All AGT levels are expressed as mean ± SE.

Isolated Limb Infusion Technique

Isolated limb infusion was done as previously described (2). Briefly, under general anesthesia, the right femoral artery and vein were isolated. The proximal femoral artery and vein were then ligated. The femoral artery was cannulated and the arterial catheter was attached to a peristaltic pump. The femoral vein was then cannulated. Once both cannulas were in place, the pump and tubing were primed with saline, the thigh tourniquet was tightened, and saline was initiated at a flow rate of 1.5 mL/min. After ensuring an appropriate flow, the limb was infused with the solution of interest. A 15-minute infusion was done at a flow rate of 1.5 mL/min, followed by a 1-minute wash-out infusion with saline at a flow rate of 3.0 mL/min. Following the wash, the arterial and venous cannulas were removed and the femoral artery and vein were ligated.

Temozolomide Treatment in the Absence and Presence of O6BG

We have previously reported a dose-escalation study using regional temozolomide (350, 750, 1,000, 1,500, and 3,000 mg/kg) in the same animal model used in this study (2). In the present study, temozolomide was administered as a single dosage via isolated limb infusion at the doses of 350 or 750 mg/kg infused limb weight. Ten percent of the rat body weight was assumed as the limb weight.

We prepared fresh temozolomide stock solution of 4 mg/mL in 10% DMSO in PBS. Standard formulations of temozolomide were kindly provided by Dr. W. Robert Bishop (Schering-Plough Research Institute, Kenilworth, NJ). Infusate was made by further dilution of temozolomide stock solution into 22.5 mL of 10% DMSO in PBS according to the given dosing regimen for a 15-minute infusion.

Modulation on AGT activity was done with i.p. O6BG following the results of O6BG dosing study. Doses of O6BG were prepared in the same manner as described above.

Once tumors reached 12 mm in greatest dimension, rats were randomly assigned to one of the following subgroups:

  • (1) Isolated limb infusion without modulation of AGT

    • Isolated limb infusion with 10% DMSO

    • Isolated limb infusion with 350 mg/kg temozolomide

    • Isolated limb infusion with 750 mg/kg temozolomide

  • (2) Isolated limb infusion in combination with O6BG

    • Isolated limb infusion with 350 mg/kg temozolomide

    • Isolated limb infusion with 750 mg/kg temozolomide

  • (3) Isolated limb infusion in combination with O6BG every 24 hours for 5 consecutive days (A total amount of O6BG per rat was 17.5 mg/kg.)

    • Isolated limb infusion with 10% DMSO

    • Isolated limb infusion with 350 mg/kg temozolomide

    • Isolated limb infusion with 750 mg/kg temozolomide

  • (4) Systemic temozolomide in combination with O6BG every 24 hours for 5 consecutive days

    • Isolated limb infusion following O6BG administration was introduced two hours after the dosing.

Tumor Measurement and End Point

Tumors and any ulcer or full-thickness eschar were measured as described above. Tumor volume was calculated according to the same formula mentioned above and the volume of any ulcer or eschar was subtracted from the total tumor volume. Animals were euthanized either when a 500% change in tumor volume occurred or after 60 days.

Evaluation of Tumor Response and Toxicity

Time (in days) taken to reach a quintupling of tumor volume was calculated from tumor volume measurements using linear regression. The average tumor quintupling was then calculated and compared. Response to treatment was also assessed by growth delays and number of regressions. Growth delay (in days) was defined as the difference in tumor quintupling time in treated versus control animals. Tumor regression was defined as a tumor of which the volume decreased over two consecutive measurements (8).

Rats were also monitored for general well-being, weight, and functional ability of leg following surgery to access the toxicity. Intra-animal weight gain or loss was calculated as a percentage of its baseline weight before surgery (day 0), and the mean nadir weight loss was calculated for each group. Toxicity was defined by assigning each rat a toxicity score as follows: grade 1, no subjective or objective evidence of reaction (normal walking pattern); grade 2, slight erythemia and/or edema; grade 3, considerable erythemia and/or edema (rodent does not use its hind limb in a useful manner but stands on it when rising); grade 4, obvious damage to the deep tissues (rat drags its hind limb without any function); grade 5, reaction that leads to autoamputation; and grade 6, animal death.

Statistical Analysis

The therapeutic indexes of the various treatments were assessed by ANOVA. When significant differences were detected in ANOVA, differences between means were checked by Bonferroni's method. Fischer's exact test was done for tumor regressions. P < 0.05 was considered significant.

Results

O6BG Dosing Study

The activity of AGT before and after exposure to O6BG is shown in Fig. 1 . The control values show AGT levels after treatment with 40% PEG in PBS, which was the vehicle used to solubilize O6BG. These values are no different than AGT values seen in the tumors of untreated animals (data not shown). Complete depletion of AGT activity in both tumors (P < 0.01) and muscle tissues (P < 0.05) within 2 to 3 hours was achieved by the administration of 17.5 and 35 mg/kg O6BG. Use of a low dose of O6BG (3.5 mg/kg) was also effective and suppressed the activity of AGT in tumors by 93.5% (P < 0.01). All rats that received 70 mg/kg O6BG and one of the three rats treated with 35 mg/kg O6BG died before tissue harvest. In view of the toxicity at higher dosing, the lowest dose tested that showed efficacy (3.5 mg/kg) was used in the remaining survival studies with only the frequency of dosing being changed.

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

AGT inhibition by O6BG in tumors and muscle tissues. Tumor and muscle AGT levels were measured in four groups of rats by giving 3.5, 17.5, 35, or 70 mg/kg O6BG i.p. (n = 3 in each group). Two to three hours after the administration, samples were harvested and used for AGT determination. The administration of 17.5 and 35 mg/kg O6BG showed complete depletion of AGT activity in both tumors and muscle tissues. A low dose of O6BG (3.5 mg/kg) also suppressed the activity of AGT in tumors by 93.5%; however, it did not significantly inhibit the activity of AGT in muscle tissues. Bars, SE.

Improvement of Therapeutic Index with Temozolomide in Conjunction with O6BG

Regional Temozolomide without O6BG. Regional temozolomide at a concentration of 350 mg/kg without modulation of AGT was ineffective at delaying tumor growth although a small increase in the number of regressions compared with the group treated with regional 10% DMSO in PBS (Table 1 ) was seen. Using a dose of 750 mg/kg regional temozolomide alone, a slight increase in quintupling time was observed, which was not significantly different from the 10% DMSO control.

View this table:
  • View inline
  • View popup
Table 1.

Effects of temozolomide and O6BG on DM6 melanoma xenografts

Regional Temozolomide with Single Dose of O6BG. Using a dose of 350 mg/kg regional temozolomide in conjunction with a single dose of 3.5 mg/kg O6BG showed increases in tumor growth delay and quintupling time that did not reach statistical significance. No increase in the number of regressions was seen over temozolomide alone (Fig. 2A ; Table 1). However, 750 mg/kg of regionally administered temozolomide with a single dose of O6BG systemically led to a significant prolongation of tumor growth delay (P < 0.01; Fig. 2B) and increased number of tumor regressions (P < 0.05; Table 1).

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Effect of regional or systemic temozolomide with or without O6BG. The average tumor growth inhibition with systemic 35 mg/kg temozolomide with 3.5 mg/kg O6BG for 5 consecutive days and intra-arterial administration of 350 mg/kg temozolomide with or without O6BG (A) and intra-arterial 10% DMSO (control) or 750 mg/kg of intra-arterial administration of temozolomide with or without O6BG (B). Y axis, percent increase in tumor volume; X axis, days after treatment. Bars, SE.

Regional Temozolomide with Multi-Dose of O6BG. When O6BG was administered systemically for 5 consecutive days, rats treated using regional isolated limb infusion with 350 or 750 mg/kg temozolomide showed significant prolongation of tumor growth delay (P < 0.01; Fig. 2A and B) and increased number of tumor regressions (P < 0.01; Table 1) compared with the group treated using isolated limb infusion with 10% DMSO in conjunction with the multi-dose course of O6BG for 5 days. In contrast, animals who received systemic temozolomide combined with multiple dosages of O6BG systemically showed no significant tumor growth delay (Table 1). Finally, to confirm that the observed activity of O6BG was not related to the vehicle (40% PEG in PBS) in which it was mixed, a series of experiments was also carried out using vehicle alone as a systemic modulator. As shown in Fig. 2B and Table 1, there is no difference in the antitumor activity of temozolomide when given in the presence or absence of 40% PEG in PBS.

Evaluation of Toxicity. As shown in Table 2 , no significant difference was found among groups in mean nadir weight loss or in maximum weight gain who underwent regional therapy using intra-arterial temozolomide in conjunction with systemic O6BG. There were no rats who died or developed an autoamputation secondary to drug toxicity using combination therapy. The group which received systemic temozolomide in conjunction with multi-dose of O6BG for 5 days showed the most severe mean nadir weight loss.

View this table:
  • View inline
  • View popup
Table 2.

Adverse effects of temozolomide and O6BG on body weight

Discussion

In a quest for new regional chemotherapeutic agents against advanced extremity melanoma, we started to explore temozolomide (temozolomide), which is a novel methylating agent and is one of the more effective single-agent therapies for metastatic melanoma (9, 10). However, because its i.v. formulation is not currently available, temozolomide has not been tried as a regional chemotherapy in humans. We initially examined dose-escalation studies using regional and systemic temozolomide in a clinically relevant animal model of regionally advanced melanoma. We found that regional temozolomide had an antitumor effect as its dose was increased whereas systemic temozolomide had little antitumor effect even at a dose exceeding the LD10 in this model (2). Furthermore, when we compared the highest equitoxic dosages of regional melphalan, a gold standard drug in regional therapy, without heat, to regional temozolomide without heat, we found that temozolomide was significantly more effective in causing regressions and delaying tumor growth than melphalan (2). These findings suggest that temozolomide might be a promising new regional therapeutic agent.

The antitumor activity of temozolomide is a direct consequence of DNA methylation. The cytotoxic lesion is felt to be due to methylation of the O6 position of guanine. Unrepaired O6-methylguanine (O6MG) seems as an “adenine” base to the cell and it is paired with a thymine base (11). The function of a repair protein, AGT, in normal cells (as well as in tumor cells) involves removing chloroethylation or methylation damage from the O6-position of guanine. If AGT is present during the repair, O6MG bases are removed and DNA repair results in the correction of the mispairing that occurred due to the methylating agent (12). However, if AGT is not present, O6MG:T mispaired bases trigger the process of DNA mismatch repair (13); then this repair process leads to a repetitive cycle of futile mismatch repair where thymine is repetitively mispaired with O6MG and subsequently leads to growth arrest and apoptosis (14, 15).

A series of laboratory studies has shown that at least two mechanisms of resistance seem to be operational in mediating resistance to temozolomide. The first of these mechanisms involves removal of the methyl adduct on the O6-position of O6MG by AGT, which has been shown in both cell culture and xenograft studies to produce resistance to temozolomide (16–20). The second mechanism of resistance is related to a deficiency in DNA mismatch repair. This mechanism has been shown to confer resistance to temozolomide in vitro (21) and in vivo (22). The futile cycle created by the inability of the DNA mismatch repair proteins to correctly fix the methyl adducts is needed for methylating agents to mediate their cytotoxic effect. The deficiency of this DNA mismatch repair process results in tolerance of O6MG residues as well as other methyl adducts in DNA and, eventually, cell survival in the presence of persistent DNA damage. Additional mechanisms of resistance to temozolomide may be operational with recent work suggesting a role for enhanced DNA base excision repair (23–25) and alterations in cell signaling involving the apoptotic pathway (26, 27).

The presence of chemotherapy resistance pathways is associated with poor tumor responses and patient survival in a variety of tumors. In melanoma cells, increased but variable AGT activity is found (28–31). There is higher AGT activity found in tumor cells than in normal skin cells and higher activity in metastatic tumors than in primary lesions. In addition, expression of AGT activity is higher in melanoma metastasis after chemotherapy with alkylating agents (28, 32).

Depletion of AGT, either with methylating agents that generate O6MG residues in DNA (33, 34) or with O6-alkylguanine (16, 35–41), renders cells more sensitive to methylator-induced cytotoxicity, presumably by way of increased formation of interstrand cross-links or initiation of a lethal cycle of mismatch repair. Dolan et al. (39, 40, 42) have reported that the substrate analogue O6-benzylguanine (O6BG) inhibits AGT activity and potentiates the cytotoxicity of both chloroethylating agents and methylating agents. They showed that O6BG binds AGT, transferring the benzyl moiety to the active-site cysteine (43). The reaction is very rapid and complete, making O6BG one of the strongest inhibitors of AGT. O6BG is not incorporated into DNA in living cells and reacts directly with both cytoplasmic and nuclear AGT. O6BG is now being used in clinical studies to assess the ability of AGT inhibition to overcome chemotherapy resistance in a variety of tumors (44–46).

When O6BG was given at concentrations of 10 to 30 mg/kg in mice, it has been reported that human tumor xenograft AGT is depleted within 30 minutes and depletion is maintained for 6 to 8 hours, after which endogenous regeneration of AGT occurs through synthesis of new protein (47, 48). In the present study, to determine an appropriate dose of O6BG to maximize AGT depletion with less toxicity in this model, we conducted O6BG dosing studies. The activity of AGT was markedly inhibited using a concentration of 3.5 mg/kg O6BG. Repeated dosing of O6BG has been shown to maintain depletion of AGT activity in tumors (49).

AGT depletion by O6BG has been shown to increase the cytotoxicity of temozolomide in vitro (50) and in vivo (6, 49, 51). Wedge et al. (49) have shown that the combination of the two agents given systemically can result in an increased therapeutic index for temozolomide in a human xenograft model. However, there is no significant difference between the groups with or without treatment of O6BG, even in the most pronounced effect in combination with 100 mg/kg systemic temozolomide. Whereas O6BG is known to inhibit AGT activity and, in several animal models, improves temozolomide efficacy in vivo against melanoma, the magnitude of the action of temozolomide is frequently limited by the synergistic toxicity that occurs when both agents are given systemically.

No previous studies have been done in the setting of regional temozolomide administration using systemic O6BG to inhibit AGT. Therefore, we then did a set of infusional treatments using two different temozolomide dosages and found marked prolongation of melanoma tumor quintupling time, increased tumor growth delay, and tumor regressions at both temozolomide dosages as compared with control treatments in animals not receiving O6BG, as well as in animals who received O6BG but had a sham (DMSO) infusion as shown in Table 1. Interestingly, there was no increase in limb toxicity seen in these animals.

In this report, we showed how the effect of regional temozolomide on an in vivo model having advanced extremity melanoma can be optimized using chemomodulation with O6BG. Modulation of chemoresistance pathways improves the therapeutic index of regional temozolomide-based therapy in our animal model of isolated limb infusion. All of these studies were done without the addition of tumor hyperthermia. These observations further support our earlier findings, using melphalan, that modulation of the tumor resistance pathways for chemotherapies used in regional infusional treatments is an effective novel strategy to improve the therapeutic index of this treatment approach (5). Our findings may offer a new approach to improve the efficacy of treatment for melanoma patients with regionally advanced disease and pave the way for a series of temozolomide-based regional therapy clinical trials.

Acknowledgments

We thank Gerald Olson, D.V.M, M.S., and Rick Leary.

Footnotes

  • Grant support: Veterans Affairs Merit Review (D.S. Tyler), Veterans Affairs Institute of Medical Research (D.S. Tyler), and Correlative Science Committee of the American College of Surgeons Oncology Group (D.S. Tyler).

  • The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

    • Accepted January 11, 2006.
    • Received April 4, 2005.
    • Revision received December 4, 2005.
  • American Association for Cancer Research

References

  1. ↵
    Jaques DP, Coit DG, Brennan MF. Major amputation for advanced malignant melanoma. Surg Gynecol Obstet 1989;169:1–6.
    OpenUrlPubMed
  2. ↵
    Ueno T, Ko SH, Grubbs E, Pruitt SK, Friedman HS, Tyler DS. Temozolomide is a novel regional infusion agent for the treatment of advanced extremity melanoma. Am J Surg 2004;188:532–7.
    OpenUrlCrossRefPubMed
  3. ↵
    Grubbs EG, Abdel-Wahab O, Cheng TY, et al. In-transit melanoma: the role of alkylating-agent resistance in regional therapy. J Am Coll Surg 2004;199:419–27.
    OpenUrlCrossRefPubMed
  4. Grubbs EG, Ueno T, Abdel-Wahab O, et al. Modulation of resistance to regional chemotherapy in the extremity melanoma model. Surgery 2004;136:210–8.
    OpenUrlCrossRefPubMed
  5. ↵
    Abdel-Wahab OI, Grubbs E, Viglianti BL, et al. The role of hyperthermia in regional alkylating agent chemotherapy. Clin Cancer Res 2004;10:5919–29.
    OpenUrlAbstract/FREE Full Text
  6. ↵
    Friedman HS, Dolan ME, Pegg AE, et al. Activity of temozolomide in the treatment of central nervous system tumor xenografts. Cancer Res 1995;55:2853–7.
    OpenUrlAbstract/FREE Full Text
  7. ↵
    Domoradzki J, Pegg AE, Dolan ME, Maher VM, McCormick JJ. Correlation between O6-methylguanine-DNA-methyltransferase activity and resistance of human cells to the cytotoxic and mutagenic effect of N-methyl-N′-nitro-N-nitrosoguanidine. Carcinogenesis 1984;5:1641–7.
    OpenUrlAbstract/FREE Full Text
  8. ↵
    Friedman HS, Keir S, Pegg AE, et al. O6-Benzylguanine-mediated enhancement of chemotherapy. Mol Cancer Ther 2002;1:943–8.
    OpenUrlAbstract/FREE Full Text
  9. ↵
    Middleton MR, Grob JJ, Aaronson N, et al. Randomized phase III study of temozolomide versus dacarbazine in the treatment of patients with advanced metastatic malignant melanoma. J Clin Oncol 2000;18:158–66.
    OpenUrlAbstract/FREE Full Text
  10. ↵
    Agarwala SS, Kirkwood JM, Gore M, et al. Temozolomide for the treatment of brain metastases associated with metastatic melanoma: a phase II study. J Clin Oncol 2004;22:2101–7.
    OpenUrlAbstract/FREE Full Text
  11. ↵
    Pauly GT, Hughes SH, Moschel RC. Response of repair-competent and repair-deficient Escherichia coli to three O6-substituted guanines and involvement of methyl-directed mismatch repair in the processing of O6-methylguanine residues. Biochemistry 1994;33:9169–77.
    OpenUrlCrossRefPubMed
  12. ↵
    Armstrong MJ, Galloway SM. Mismatch repair provokes chromosome aberrations in hamster cells treated with methylating agents or 6-thioguanine, but not with ethylating agents. Mutat Res 1997;373:167–78.
    OpenUrlPubMed
  13. ↵
    Aquilina G, Hess P, Branch P, et al. A mismatch recognition defect in colon carcinoma confers DNA microsatellite instability and a mutator phenotype. Proc Natl Acad Sci U S A 1994;91:8905–9.
    OpenUrlAbstract/FREE Full Text
  14. ↵
    D'Atri S, Tentori L, Lacal PM, et al. Involvement of the mismatch repair system in temozolomide-induced apoptosis. Mol Pharmacol 1998;54:334–41.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    Hickman MJ, Samson LD. Role of DNA mismatch repair and p53 in signaling induction of apoptosis by alkylating agents. Proc Natl Acad Sci U S A 1999;96:10764–9.
    OpenUrlAbstract/FREE Full Text
  16. ↵
    Yarosh DB, Hurst-Calderone S, Babich MA, Day RS III. Inactivation of O6-methylguanine-DNA methyltransferase and sensitization of human tumor cells to killing by chloroethylnitrosourea by O6-methylguanine as a free base. Cancer Res 1986;46:1663–8.
    OpenUrlAbstract/FREE Full Text
  17. Pegg AE. Mammalian O6-alkylguanine-DNA alkyltransferase: regulation and importance in response to alkylating carcinogenic and therapeutic agents. Cancer Res 1990;50:6119–29.
    OpenUrlFREE Full Text
  18. Ludlum DB. DNA alkylation by the haloethylnitrosoureas: nature of modifications produced and their enzymatic repair or removal. Mutat Res 1990;233:117–26.
    OpenUrlCrossRefPubMed
  19. Pegg AE, Byers TL. Repair of DNA containing O6-alkylguanine. FASEB J 1992;6:2302–10.
    OpenUrlAbstract
  20. ↵
    Pegg AE, Dolan ME, Moschel RC. Structure, function, and inhibition of O6-alkylguanine-DNA alkyltransferase. Prog Nucleic Acid Res Mol Biol 1995;51:167–223.
    OpenUrlCrossRefPubMed
  21. ↵
    Liu L, Markowitz S, Gerson SL. Mismatch repair mutations override alkyltransferase in conferring resistance to temozolomide but not to 1,3-bis(2-chloroethyl)nitrosourea. Cancer Res 1996;56:5375–9.
    OpenUrlAbstract/FREE Full Text
  22. ↵
    Friedman HS, Johnson SP, Dong Q, et al. Methylator resistance mediated by mismatch repair deficiency in a glioblastoma multiforme xenograft. Cancer Res 1997;57:2933–6.
    OpenUrlAbstract/FREE Full Text
  23. ↵
    Liu L, Taverna P, Whitacre CM, Chatterjee S, Gerson SL. Pharmacologic disruption of base excision repair sensitizes mismatch repair-deficient and -proficient colon cancer cells to methylating agents. Clin Cancer Res 1999;5:2908–17.
    OpenUrlAbstract/FREE Full Text
  24. Tentori L, Leonetti C, Scarsella M, et al. Combined treatment with temozolomide and poly(ADP-ribose) polymerase inhibitor enhances survival of mice bearing hematologic malignancy at the central nervous system site. Blood 2002;99:2241–4.
    OpenUrlAbstract/FREE Full Text
  25. ↵
    Tentori L, Portarena I, Torino F, Scerrati M, Navarra P, Graziani G. Poly(ADP-ribose) polymerase inhibitor increases growth inhibition and reduces G(2)/M cell accumulation induced by temozolomide in malignant glioma cells. Glia 2002;40:44–54.
    OpenUrlCrossRefPubMed
  26. ↵
    Bocangel DB, Finkelstein S, Schold SC, Bhakat KK, Mitra S, Kokkinakis DM. Multifaceted resistance of gliomas to temozolomide. Clin Cancer Res 2002;8:2725–34.
    OpenUrlAbstract/FREE Full Text
  27. ↵
    Ma J, Murphy M, O'Dwyer PJ, Berman E, Reed K, Gallo JM. Biochemical changes associated with a multidrug-resistant phenotype of a human glioma cell line with temozolomide-acquired resistance. Biochem Pharmacol 2002;63:1219–28.
    OpenUrlCrossRefPubMed
  28. ↵
    Moriwaki S, Nishigori C, Takebe H, Imamura S. O6-Alkylguanine-DNA alkyltransferase activity in human malignant melanoma. J Dermatol Sci 1992;4:6–10.
    OpenUrlCrossRefPubMed
  29. Lee SM, Rafferty JA, Elder RH, et al. Immunohistological examination of the inter- and intracellular distribution of O6-alkylguanine DNA-alkyltransferase in human liver and melanoma. Br J Cancer 1992;66:355–60.
    OpenUrlPubMed
  30. Middleton MR, Lunn JM, Morris C, et al. O6-Methylguanine-DNA methyltransferase in pretreatment tumour biopsies as a predictor of response to temozolomide in melanoma. Br J Cancer 1998;78:1199–202.
    OpenUrlPubMed
  31. ↵
    Ma S, Egyhazi S, Martenhed G, Ringborg U, Hansson J. Analysis of O(6)-methylguanine-DNA methyltransferase in melanoma tumours in patients treated with dacarbazine-based chemotherapy. Melanoma Res 2002;12:335–42.
    OpenUrlCrossRefPubMed
  32. ↵
    Spiro T, Liu L, Gerson S. New cytotoxic agents for the treatment of metastatic malignant melanoma: temozolomide and related alkylating agents in combination with guanine analogues to abrogate drug resistance. Forum (Genova) 2000;10:274–85.
  33. ↵
    Zlotogorski C, Erickson LC. Pretreatment of human colon tumor cells with DNA methylating agents inhibits their ability to repair chloroethyl monoadducts. Carcinogenesis 1984;5:83–7.
    OpenUrlAbstract/FREE Full Text
  34. ↵
    Aida T, Bodell WJ. Cellular resistance to chloroethylnitrosoureas, nitrogen mustard, and cis-diamminedichloroplatinum(II) in human glial-derived cell lines. Cancer Res 1987;47:1361–6.
    OpenUrlAbstract/FREE Full Text
  35. ↵
    Gerson SL, Trey JE, Miller K. Potentiation of nitrosourea cytotoxicity in human leukemic cells by inactivation of O6-alkylguanine-DNA alkyltransferase. Cancer Res 1988;48:1521–7.
    OpenUrlAbstract/FREE Full Text
  36. Dolan ME, Corsico CD, Pegg AE. Exposure of HeLa cells to O(6)-alkylguanines increases sensitivity to the cytotoxic effects of alkylating agents. Biochem Biophys Res Commun 1985;132:178–85.
    OpenUrlCrossRefPubMed
  37. Dolan ME, Morimoto K, Pegg AE. Reduction of O6-alkylguanine-DNA alkyltransferase activity in HeLa cells treated with O6-alkylguanines. Cancer Res 1985;45:6413–7.
    OpenUrlAbstract/FREE Full Text
  38. Dolan ME, Young GS, Pegg AE. Effect of O6-alkylguanine pretreatment on the sensitivity of human colon tumor cells to the cytotoxic effects of chloroethylating agents. Cancer Res 1986;46:4500–4.
    OpenUrlAbstract/FREE Full Text
  39. ↵
    Dolan ME, Moschel RC, Pegg AE. Depletion of mammalian O6-alkylguanine-DNA alkyltransferase activity by O6-benzylguanine provides a means to evaluate the role of this protein in protection against carcinogenic and therapeutic alkylating agents. Proc Natl Acad Sci U S A 1990;87:5368–72.
    OpenUrlAbstract/FREE Full Text
  40. ↵
    Dolan ME, Mitchell RB, Mummert C, Moschel RC, Pegg AE. Effect of O6-benzylguanine analogues on sensitivity of human tumor cells to the cytotoxic effects of alkylating agents. Cancer Res 1991;51:3367–72.
    OpenUrlAbstract/FREE Full Text
  41. ↵
    Mitchell RB, Moschel RC, Dolan ME. Effect of O6-benzylguanine on the sensitivity of human tumor xenografts to 1,3-bis(2-chloroethyl)-1-nitrosourea and on DNA interstrand cross-link formation. Cancer Res 1992;52:1171–5.
    OpenUrlAbstract/FREE Full Text
  42. ↵
    Dolan ME, Stine L, Mitchell RB, Moschel RC, Pegg AE. Modulation of mammalian O6-alkylguanine-DNA alkyltransferase in vivo by O6-benzylguanine and its effect on the sensitivity of a human glioma tumor to 1-(2-chloroethyl)-3-(4-methylcyclohexyl)-1-nitrosourea. Cancer Commun 1990;2:371–7.
    OpenUrlPubMed
  43. ↵
    Pegg AE, Boosalis M, Samson L, et al. Mechanism of inactivation of human O6-alkylguanine-DNA alkyltransferase by O6-benzylguanine. Biochemistry 1993;32:11998–2006.
    OpenUrlCrossRefPubMed
  44. ↵
    Spiro TP, Gerson SL, Liu L, et al. O6-Benzylguanine: a clinical trial establishing the biochemical modulatory dose in tumor tissue for alkyltransferase-directed DNA repair. Cancer Res 1999;59:2402–10.
    OpenUrlAbstract/FREE Full Text
  45. Schilsky RL, Dolan ME, Bertucci D, et al. Phase I clinical trial and pharmacological study of O6-benzylguanine followed by carmustine in patients with advanced cancer. Clin Cancer Res 2000;6:3025–31.
    OpenUrlAbstract/FREE Full Text
  46. ↵
    Friedman HS, Kokkinakis DM, Pluda J, et al. Phase I trial of O6-benzylguanine for patients undergoing surgery for malignant glioma. J Clin Oncol 1998;16:3570–5.
    OpenUrlAbstract
  47. ↵
    Gerson SL, Zborowska E, Norton K, Gordon NH, Willson JK. Synergistic efficacy of O6-benzylguanine and 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) in a human colon cancer xenograft completely resistant to BCNU alone. Biochem Pharmacol 1993;45:483–91.
    OpenUrlCrossRefPubMed
  48. ↵
    Kreklau EL, Kurpad C, Williams DA, Erickson LC. Prolonged inhibition of O(6)-methylguanine DNA methyltransferase in human tumor cells by O(6)-benzylguanine in vitro and in vivo. J Pharmacol Exp Ther 1999;291:1269–75.
    OpenUrlAbstract/FREE Full Text
  49. ↵
    Wedge SR, Porteous JK, Newlands ES. Effect of single and multiple administration of an O6-benzylguanine/temozolomide combination: an evaluation in a human melanoma xenograft model. Cancer Chemother Pharmacol 1997;40:266–72.
    OpenUrlCrossRefPubMed
  50. ↵
    Wedge SR, Porteous JK, May BL, Newlands ES. Potentiation of temozolomide and BCNU cytotoxicity by O(6)-benzylguanine: a comparative study in vitro. Br J Cancer 1996;73:482–90.
    OpenUrlCrossRefPubMed
  51. ↵
    Wedge SR, Newlands ES. O6-Benzylguanine enhances the sensitivity of a glioma xenograft with low O6-alkylguanine-DNA alkyltransferase activity to temozolomide and BCNU. Br J Cancer 1996;73:1049–52.
    OpenUrlPubMed
PreviousNext
Back to top
Molecular Cancer Therapeutics: 5 (3)
March 2006
Volume 5, Issue 3
  • Table of Contents
  • About the Cover

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Molecular Cancer Therapeutics article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Modulation of chemotherapy resistance in regional therapy: a novel therapeutic approach to advanced extremity melanoma using intra-arterial temozolomide in combination with systemic O6-benzylguanine
(Your Name) has forwarded a page to you from Molecular Cancer Therapeutics
(Your Name) thought you would be interested in this article in Molecular Cancer Therapeutics.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Modulation of chemotherapy resistance in regional therapy: a novel therapeutic approach to advanced extremity melanoma using intra-arterial temozolomide in combination with systemic O6-benzylguanine
Tomio Ueno, Sae Hee Ko, Elizabeth Grubbs, Yasunori Yoshimoto, Christi Augustine, Zeinab Abdel-Wahab, Tsung-Yen Cheng, Omar I. Abdel-Wahab, Scott K. Pruitt, Henry S. Friedman and Douglas S. Tyler
Mol Cancer Ther March 1 2006 (5) (3) 732-738; DOI: 10.1158/1535-7163.MCT-05-0098

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Modulation of chemotherapy resistance in regional therapy: a novel therapeutic approach to advanced extremity melanoma using intra-arterial temozolomide in combination with systemic O6-benzylguanine
Tomio Ueno, Sae Hee Ko, Elizabeth Grubbs, Yasunori Yoshimoto, Christi Augustine, Zeinab Abdel-Wahab, Tsung-Yen Cheng, Omar I. Abdel-Wahab, Scott K. Pruitt, Henry S. Friedman and Douglas S. Tyler
Mol Cancer Ther March 1 2006 (5) (3) 732-738; DOI: 10.1158/1535-7163.MCT-05-0098
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgments
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • Prediction of individual response to platinum/paclitaxel combination using novel marker genes in ovarian cancers
  • Low doses of cisplatin or gemcitabine plus Photofrin/photodynamic therapy: Disjointed cell cycle phase-related activity accounts for synergistic outcome in metastatic non–small cell lung cancer cells (H1299)
  • Semisynthetic homoharringtonine induces apoptosis via inhibition of protein synthesis and triggers rapid myeloid cell leukemia-1 down-regulation in myeloid leukemia cells
Show more Article
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About MCT

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Molecular Cancer Therapeutics
eISSN: 1538-8514
ISSN: 1535-7163

Advertisement