Skip to main content
  • AACR Publications
    • 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
    • CCR Focus Archive
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Breast Cancer
      • Clinical Trials
      • Immunotherapy: Facts and Hopes
      • 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 Publications
    • 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
Clinical Cancer Research
Clinical Cancer Research
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • CCR Focus Archive
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Breast Cancer
      • Clinical Trials
      • Immunotherapy: Facts and Hopes
      • 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

Cancer Therapy: Clinical

Phase II Trial of Karenitecin in Patients with Malignant Melanoma: Clinical and Translational Study

Adil Daud, Nikola Valkov, Barbara Centeno, Jennifer Derderian, Patricia Sullivan, Pamela Munster, Patricia Urbas, Ronald C. DeConti, Elmer Berghorn, Zhenmei Liu, Frederick Hausheer and Daniel Sullivan
Adil Daud
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Nikola Valkov
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Barbara Centeno
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jennifer Derderian
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Patricia Sullivan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Pamela Munster
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Patricia Urbas
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Ronald C. DeConti
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Elmer Berghorn
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Zhenmei Liu
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Frederick Hausheer
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Daniel Sullivan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1158/1078-0432.CCR-04-1722 Published April 2005
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Purpose: A phase II trial of the novel camptothecin karenitecin (BNP1350) was conducted to determine its efficacy and tolerability in patients with metastatic melanoma. Patients were biopsied to determine topoisomerase expression at baseline and response to therapy.

Patients and Methods: Eligible patients had metastatic melanoma with up to three prior chemotherapy and/or any number of immunotherapy regimens. Treatment consisted of an i.v. infusion of 1 mg/m2 karenitecin daily for 5 days with cycles repeated every 3 weeks. Fine-needle aspiration biopsies were done before treatment and on day 3 to determine topoisomerase expression from patients' tumors.

Results: Forty-three patients were evaluable for response and toxicity. Most patients (72%) had stage M1C disease and were previously exposed to chemotherapy (56%). The investigational agent was well tolerated with limited gastrointestinal side effects or fatigue. The major toxicity seen was reversible noncumulative myelosuppression. One patient had a complete response after 11 months of therapy. No partial responses were seen, but 33% of the patients had disease stabilization lasting ≥3 months. Topoisomerase I, IIα, and IIβ expression and localization were determined in a subset of patients. Topoisomerase I expression was highest, followed by topoisomerase IIβ and topoisomerase IIα.

Conclusion: Karenitecin was a well-tolerated investigational agent in this phase II study; side effects were generally mild and mostly hematologic. Karenitecin has significant activity in metastatic melanoma. Melanoma metastases express high levels of topoisomerase I. We did not observe any compensatory increase in topoisomerase II upon treatment with karenitecin.

  • Topoisomerase I
  • Karenitecin
  • Melanoma

Advanced malignant melanoma with distant metastasis is a difficult disease to treat with a poor outcome in most cases. Although dacarbazine is considered the “standard of care,” it has a response rate of only 10% to 15% with a response duration of 4 to 6 months (1, 2). The addition of other chemotherapy agents and cytokines has helped response rates but not overall survival (3). The recent Eastern Cooperative Oncology Group trial (ECOG 3695) presented at American Society of Clinical Oncology 2003 conference reiterated the lack of an advantage of the biochemotherapy regimens (dacarbazine, cisplatin, vinblastine, interleukin-2, and IFN-α) over chemotherapy (dacarbazine, cisplatin, and vinblastine) alone (4). Second-line chemotherapy of patients with melanoma is even more limited in its effectiveness.

During the past decade, camptothecin analogues (topoisomerase I inhibitors) have emerged as an important new class of antitumor drugs with clinical utility in colon, ovarian, lung, and other types of cancer (5). The clinical utility of these compounds can be limited by clinical toxicity, reduced tissue diffusion, interpatient variability, and susceptibility to drug-resistance pumps (6). Further, currently available camptothecins have low concentrations of the lactone species in human plasma (≤20% of total drug concentration). The lactone species is the active form of camptothecins; if the plasma concentrations of this species could be increased, it is likely that it would result in a relative increase in antitumor activity.

Karenitecin (7-[(2-trimethylsilyl) ethyl]-20(S)-camptothecin) is a novel, lipophilic camptothecin derivative (7). It has enhanced lactone stability (∼85%) in human plasma. This compound is not a prodrug and does not undergo glucuronidation, thereby reducing the amount of interpatient variability and gastrointestinal toxicity (7). Karenitecin has shown significant activity in vitro and in vivo against various human xenograft tumor models, including melanoma. Its antitumor activity in these models is similar or superior to other camptothecins. In a phase I study, dose-limiting toxicity consisted of reversible grade 4 neutropenia and thrombocytopenia in patients treated at 1.2 and 2.4 mg/m2 (8). There was no cumulative marrow suppression. The recommended phase II dose of karenitecin was 1 mg/m2 daily for 5 consecutive days using a Q 3 week schedule. We initiated a phase II study of karenitecin in advanced melanoma to evaluate activity in this patient population and to study the relationship of topoisomerase expression in melanoma to response. Because the role of topoisomerase expression in response to therapy is currently not clear, we also characterized topoisomerase expression in patients with tumors that can be biopsied before and during treatment.

Patients and Methods

Enrollment criteria. A phase II trial of karenitecin (MCC 12824) was initiated at our institution on March 2002. Patient enrollment ended April 2003 once target enrollment was completed. The study design was approved by the Scientific Review Committee at Moffitt Cancer Center and the University of South Florida Institutional Review Board. Informed consent was obtained from all patients. Patients were required to have histologically or cytologically proven metastatic melanoma with measurable disease, and an ECOG performance status of 0 or 1. Patients could have had up to three prior chemotherapy regimens and central nervous system disease if the disease was stable or resected. Skin, mucosal, and uveal primary sites were eligible. Patients were excluded from this study if they were pregnant or nursing or had an active second malignancy. Patients were excluded if the following initial laboratory parameters were abnormal (bilirubin >1.5, creatinine <1.4 ANC <1,500, or platelets <100,000).

Statistical design and response assessment. This study was designed as an open-label, single-institution phase II trial to test the efficacy of karenitecin in melanoma. A Simon two-stage optimal trial design was followed. We planned to accrue 12 patients in the first stage of the trial and to close accrual if no response was seen (defined as a complete or partial response or as stable disease of ≥3 months duration). If one or more responses were seen, we would enroll up to 45 evaluable patients. We estimated that if this treatment had a 25% true response rate, we would have a 3% chance of stopping the study early and a 81% chance of considering it worthy of further study. At the end of stage 2, if seven or more responses of the 45 patients were seen, then the treatment would be considered worthy of further study. Bidimensional measurement (sum of the product of longest perpendicular diameters) was used to determine response/progression. A complete response was defined as resolution of all disease lesions with no new lesions and normalization of any elevated tumor markers confirmed at least 4 weeks after the initial observation. Partial response was defined as 50% reduction in bidimensional measurements confirmed at least 4 weeks after the first observed reduction. Minor response was defined as 25% to 49% reduction in bidimensional measurement. Stable disease was defined as neither shrinkage sufficient to qualify for partial response/complete response/minor response nor increase sufficient to qualify for progressive disease. Progressive disease was defined as increase in bidimensional measurement by 25%. Clinical benefit included patients with complete response, partial response, and stable disease for at least 3 months.

Treatment. All patients received 1 mg/m2/d karenitecin for 5 days consecutively. Treatment cycles were repeated every 21 days. Patients were premedicated with 8 mg ondansetron and 10 mg dexamethasone IV. The dexamethasone premedication was dropped in subsequent cycles if little or no nausea was observed. No colony stimulating factor support was utilized in this study; erythropoietin could be used for symptomatic anemia with a hemoglobin <10 g/dL and for any anemia with a hemoglobin <8 g/dL. Patients were evaluated for disease progression every two cycles. Treatment was continued only if there was stable disease (<25% increase in disease) or if there was disease reduction as evaluated by bidimensional measurements on computed tomography scan or on physical exam. Dose reductions for future cycles were based on hematologic or hepatic dysfunction; for febrile neutropenia, dose was reduced by 25% in succeeding cycles. If counts had not recovered to at least ANC ≥1,500 or platelets ≥100,000, treatment was held for 1 or 2 weeks until recovery of counts. If patients had two consecutive cycles of delay, the dose was reduced to 0.75 mg/m2 and then to 0.5 mg/m2.

Topoisomerase assay and fine-needle aspiration biopsy. Patients were consented to have biopsy only if they had metastases that can be biopsied with minimal risk. The biopsy was attempted on day 1 before the start of therapy and then on day 3 of the first cycle of treatment. Biopsy was done with a 23-gauge needle and two to four passes were done on each lesion by the study pathologist (Dr. B. Centeno). Aspirated cells were stained immediately in Diff-Quick and examined under a microscope to verify that a pure population of melanoma tumor cells was obtained. A sample was aspirated into 0.9% saline, serial cytospin slides were prepared, and frozen at −20°C until assayed. Quantitative immunflourescent staining for topoisomerase I, IIα, and IIβ was done as described previously (9). Briefly, slides were thawed and fixed at 20°C with 4% paraformaldehyde in PBS (Electron Microscopy Sciences, Fort Washington, PA) for 15 minutes. The cross-linking was stopped by rinsing slides in 1% glycine in PBS and cells were permeabilized for 24 hours by several changes of 0.25% Triton X-100 in 1% glycine-PBS. Rabbit polyclonal antibody 454 against topoisomerase IIα was combined with antihistone antibody diluted 1:100 with 0.1% NP40, 1% bovine serum albumin in PBS, and incubated for 1 hour at room temperature with the primary antibodies. The other combination included monoclonal anti–topoisomerase I and polyclonal anti–topoisomerase IIβ antibody. After three washes with PBS for 1 hour, the slides were incubated with 1:80 diluted goat anti-rabbit IgG-tetramethylrhodamine isothiocyanate labeled for the polyclonal primary antibodies and 1:125 diluted goat anti-mouse IgG-FITC for the monoclonal antibodies (Sigma, St. Louis, MO) in 0.1% NP40 and 1% bovine serum albumin in PBS for 35 minutes at room temperature. After three washes in PBS, the slides were dried and covered with coverslips in Vectashield mounting media of antifade/4′,6-diamidino-2-phenylindole (1:1; Vector Laboratories, Inc., Burlingame, CA). Immunofluorescence was observed with a Leitz Orthoplan 2 microscope and images were captured by a charged coupled device camera with Smart Capture program (Vysis, Downers Grove, IL). Quantification was done using the pixel count of the image obtained and was reported in arbitrary units.

Results and Analyses

Software package SAS version 8.2 was used in all of the following statistical analyses.

Patient characteristics. A total of 46 patients were enrolled on trial MCC 12824. One patient withdrew consent during his initial treatment. Another two patients were found to be ineligible for treatment due to violation of entry criteria (ECOG performance status >2). An additional two patients were not evaluable as they had stable disease after two cycles but decided not to continue on treatment. Therefore, a total of 41 patients were evaluable for response, 43 were evaluable for toxicity, and 45 patients were evaluable for survival. Patient characteristics are described in Table 1. The age, sex, and race distribution are close to our institutional and national norms. The majority of patients had visceral involvement and/or elevated lactate dehydrogenase; 72% of patients had stage M1C disease. There was a significant number of patients treated who had nonconjunctival ocular (6 of 41) and mucosal (2 of 41) primaries. Fifty-six percent of patients had previously received chemotherapy (mostly DTIC based, including biochemotherapy), and 26% had failed two prior chemotherapy regimens. Twenty-six patients had received radiation therapy, and 5 of 43 patients had brain metastases, which were “stable” before the start of therapy.

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

Demographic characteristics of patients

Treatment responses. In this trial, we had a total of 41 patients evaluable for response and 14 responses (complete response + partial response + minor response + stable disease). Estimated response rate is 0.341, with a SE of 0.074. Details of response of patients to this treatment are shown in Tables 2 and 3. One patient experienced a complete response to treatment. He had received 16 cycles of treatment with regression of skin lesions after the first four cycles and developed stable necrotic appearing lesions in the pancreas on computed tomography scan. These lesions showed no uptake on positron emission tomography scan and were resected. Pathology from resected lesions showed intrapancreatic and peripancreatic lymph nodes completely replaced with necrotic tumor (Fig. 1). Interestingly, this patient had previously progressed on concurrent biochemotherapy (cisplatin, vinblastine, dacarbazine, interleukin 2, and IFN-I) and then on an investigational trial with Epothilone B (Epo 908, Novartis, East Hanover, NJ). A significant number of patients (32%) or 13 of 41 experienced minor responses or disease stabilization (Tables 2-4). The longest to date is a patient with ocular melanoma with bone, brain, liver, and soft tissue metastatases had stable disease lasting for 12 cycles (36 weeks). He had stable brain metastasis on karenitecin for 36 weeks without any other treatment during this entire period. His treatments were held as he needed surgery for an unrelated condition and he progressed soon afterward. Several other patients had relatively long periods of stable disease. Patients with ocular melanoma particularly appeared to benefit from karenitecin; three of five patients with ocular melanoma had stable disease. Of these, one patient had previous chemoembolization of liver lesions with progression of disease before treatment and remained stable for 18 weeks on karenitecin; one patient had previous radiation therapy and remained stable for 36 weeks (see above); and one patient who had no prior therapy was diagnosed with concurrent ocular and liver disease and remained stable for 18 weeks on therapy.

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

Response to karenitecin therapy

View this table:
  • View inline
  • View popup
Table 3.

Characteristics of responders

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

Patient SAB 4 was treated with karenitecin for 16 cycles (48 weeks). He had regression of subcutaneous disease with stable intra-abdominal disease by computed tomography scan. He had a positron emission tomography scan showing no glucose uptake (data not shown). He underwent resection of two intrapancreatic and peripancreatic lesions. Left, a low-power view of an intrapancreatic lymph node replaced with metastatic malignant melanoma that showed nearly 100% necrosis. Right, central region at high power showing melanotic cellular debris and ghost-like remnants of tumor cells.

View this table:
  • View inline
  • View popup
Table 4.

Adverse effects observed in patients

Survival data analysis. Forty-five patients were evaluable for survival. Progression-free survival was computed as the time between beginning treatment and disease progression (Fig. 2). Overall survival time was defined as the time beginning at treatment until death, last known follow-up, or off treatment (Fig. 3). Kaplan-Meier curves were used to describe progression-free survival and overall survival. The median progression-free survival time was 7.9 weeks and corresponding 95% confidence limit was (7.0, 9.1) weeks. The Kaplan-Meier progression-free survival curve for all evaluable patients is shown in Fig. 2.

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

The Kaplan-Meier progression-free survival curve for all evaluable patients treated with karenitecin.

Fig. 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 3.

The Kaplan-Meier overall survival curve for all evaluable patients treated with karenitecin.

For overall survival, the median was 33.4 weeks and corresponding 95% confidence interval was (21.0, 38.7) weeks. The percentage of the patients that are still living is 26.67%. The Kaplan-Meier overall survival curve for all evaluable patients is shown in Fig. 3.

Toxicity. Karenitecin was well tolerated in this trial. Adverse effects seen on therapy are shown in Table 4. The predominant toxicity was hematologic. Anemia was the most frequent toxicity (79% of patients experiencing some degree of anemia) with 19% of patients experiencing grade 3 anemia. Neutropenia and thrombocytopenia were also relatively frequent (44% each). We did not use growth factor support during this trial and in most patients neutropenia resolved with dose delay. Thirty-two percent of patients experienced either grade 3 or 4 neutropenia. Neither neutropenia nor thrombocytopenia seemed to be cumulative. One patient died while on treatment with an intracranial hemorrhage that occurred during the platelet nadir (this patient was also taking aspirin). While gastrointestinal symptoms had been anticipated, only mild abdominal pain and nausea were seen with any frequency. The overall incidence of diarrhea (only grade 1 and 2 were seen) was 16%. Local infusional site discomfort was a notable complaint in 18% of patients and a sterile phlebitis developed in three patients. Fatigue (usually mild and transient) was also frequently noted.

Assessment of tumor specimen topoisomerase levels by immunofluorescence. The fine needle aspiration biopsy sample provided an abundant (>100,000 cells per sample) and nearly homogenous (>95% pure) population of melanoma cells (Fig. 4A) for immunostaining. Thirteen patients had pretreatment and posttreatment biopsies; a further seven patients had only pretreatment biopsies. Abundant expression of topoisomerase I was seen in patients who underwent biopsy (Figs. 4 and 5). We also profiled topoisomerase IIα and IIβ expression and location, which have been reported to increase following topoisomerase I inhibitor treatment (10). We found no increase on day 3 of treatment compared with pretreatment levels; we do report high levels of these enzymes, especially topoisomerase IIβ in most melanoma cells (Fig. 5). No relationship was seen between pretreatment topoisomerase I expression and response to therapy or to localization with karenitecin treatment.

Fig. 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 4.

Topoisomerase I, IIα, and IIβ expression in melanoma cells. Fine-needle aspiration was done immediately before treatment and on day 3 of treatment with karenitecin. Aspirated cells were stained to verify that a pure population of melanoma tumor cells was obtained (A). Immunoflourescent staining for topoisomerase I, IIα, and IIβ, respectively, was done (B). Western blotting was also done (bottom). Controls are HL-60 cells at days 5, 2.5, and 1 after differentiation, and melanoma cells are shown at day 1 pretreatment and day 3 of treatment (d3).

Fig. 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 5.

Cumulative topoisomerase I, IIα, and IIβ expressions in nuclei of melanoma cells. Bars, SE.

Discussion

Current therapy for metastatic melanoma remains highly unsatisfactory (1, 11, 12). Most chemotherapy regimens contain the alkylating agent dacarbazine. This drug and its congener, temozolomide, produce objective response rates in patients with melanoma in 10% to 20% of patients. It is currently not known if dacarbazine prolongs survival in patients with melanoma. Combination chemotherapy in the form of the “Dartmouth regimen” has higher reported response rates, but in a prospective randomized trial showed no advantage in survival over dacarbazine alone (13). More recently, IFN-α and interleukin-2 have been incorporated into biochemotherapy regimens that also showed very high response rates (45-60%) in single-institution phase II trials. However, in a prospective randomized clinical trial, biochemotherapy failed to show any survival benefit over chemotherapy alone (4). Therefore, it seems that varying combinations of currently used drugs will have only limited utility in this disease.

The topoisomerase I inhibitors have shown a wide spectrum of activity in human malignancies. Whereas initial studies of sodium camptothecin, which was formulated in sodium hydroxide that lead to hydrolysis of the E-ring lactone, had limited clinical utility due to its severe side effect profile (especially hemorrhagic cystitis) and absence of antitumor activity (probably because the drug was in the carboxylate form), other topoisomerase I inhibitors, such as topotecan and irinotecan, have proved useful in the clinic. Two other trials of the topoisomerase I inhibitor topotecan have been reported to our knowledge (14, 15). In the first study, 28 patients with melanoma were treated, 27 were evaluable, and 1 patient with M1a disease had a partial response. No statistics were reported on stable disease. In the second study, presented in abstract form only (15), 17 patients were treated in this study and no activity was seen. The major side effect seen was myelosuppression (severe grade 3-4 in 70% of this small sample). Topoisomerase II inhibitors have also been explored in melanoma. Epirubicin was tested in several phase II trials and not found to be effective (16–18). In our preliminary analysis, melanoma cells expressed high levels of topoisomerase I. Furthermore, we found that karenitecin, a novel camptothecin analogue, showed activity against melanoma cell lines in vitro. This molecule has a novel silyl substitution in the camptothecin B-ring, which enhances the drug's lipophilicity, increases its lactone stability, and the agent cannot undergo glucuronidation, which, in turn, reduces intrapatient variability, and the agent has the lowest observed susceptibility to breast cancer resistance protein–mediated drug resistance (19, 20). Although not clinically proven, based on its liphophilic nature, karenitecin seems to have some degree of central nervous system penetration, which would be a major advantage in melanoma with its high proportion of central nervous system involvement.

We found in the current trial that karenitecin has appreciable activity in melanoma. Even in the poorest prognosis patients (visceral disease with prior chemotherapy treatment), it was possible to have prolonged disease stabilization. Indeed, the only patient who experienced a complete response on this trial had 16 cycles of therapy after having progressed on biochemotherapy and on an investigational trial of Epothilone B. Having said that, the major treatment effect we observed in this trial was disease stabilization rather than tumor shrinkage. Some of the disease stabilization was accompanied by a decrease in lactate dehydrogenase for the duration of treatment (the longest being 9 months). Again, this effect was observed both in patients with progression on previous regimens and on previously untreated patients. Another interesting aspect of this study was the inclusion of patients with nonconjunctival ocular melanoma and mucosal melanoma. In patients with ocular melanoma 60% (three of five patients) had disease stabilization. Whereas this sample is too small to draw any conclusions, they do deserve to be included in future trials of novel agents in melanoma and in light of their biological differences with cutaneous melanoma may actually have better responses to treatment.

Treatment with karenitecin was generally well tolerated. The major complaint voiced by patients was the inconvenience of having treatment 5 of 21 days resulting in a disruption of lifestyles. The major side effect observed in this study was noncumulative reversible hematologic toxicity. Twelve of 38 patients had grade 3 or 4 neutropenia. Because no growth factor support was allowed, dose delays were the major consequence of this toxicity. Anemia was also frequent with 87% of patients having some anemia and 23% having moderate or severe (grade 3 or 4) anemia. Thrombocytopenia occurred in 50% of patients treated and six patients had moderate to severe thrombocytopenia. The one death on the study that was considered related to treatment occurred in a patient who had had severe thrombocytopenia in a previous cycle and had a bleed related to a brain metastasis.

There is very limited prospective human data on topoisomerase expression. One of the major aims of this study was to profile topoisomerase expression in melanoma before and during treatment. In preclinical studies, topoisomerase IIα levels have been shown to increase with topoisomerase I inhibitor treatment (10). We found no significant change of topoisomerase IIα with karenitecin treatment. The discrepancy may relate to the tumor type studied or due to a difference between the model studied by Whitacre et al. and human subjects studied in our trial. Previous studies have suggested that topoisomerase expression may correlate to sensitivity to topoisomerase poisons (21–23). Other studies have not shown a correlation between topoisomerase expression and outcome (24, 25). One caveat with these studies is that determination of topoisomerase expression in paraffin-embedded blocks may allow translocation of topoisomerase and, therefore, make it hard to evaluate expression, this may be reduced by rapid fixation of live cells, such as those used in our study (9). Another concern with topoisomerase expression has been evaluation of nuclear topoisomerase as opposed to total cellular topoisomerase (which is the active enzyme; refs. 26–29). Therefore, we quantified topoisomerase expression in nuclei taking precautions to reduce processing time. Because topoisomerase treatment could reduce levels of topoisomerase, we sampled tumors both before and on day 3 of treatment (30, 31). Additionally, we measured topoisomerase isoforms that are not targets of karenitecin to evaluate if any compensatory change in expression may occur during treatment 10. We found abundant expression of topoisomerase in the majority of tumors examined (Fig. 4). There was especially high expression of topoisomerase I, suggesting that this may be an interesting target for therapy in melanoma. Most of the topoisomerase expression was nuclear, rather than cytoplasmic, suggesting its utility. Topoisomerase I expression did increase (although this was not significant) with treatment (Figs. 4 and 5). Additionally, high level expression of topoisomerase IIβ was also seen, suggesting that this may be an interesting target for future trials. We did not find evidence of nuclear to cytoplasmic translocation of topoisomerase isoforms, which has been reported as a potential mechanism of resistance to topoisomerase poisons. Because only easily accessible tumors were biopsied and most patients with clinical benefit tended not to have tumors that can easily be biopsied, only three patients with clinical benefit had topoisomerase expression profiling. Hence, no conclusions could be drawn regarding topoisomerase expression and/or localization and response. Another caveat to these analyses is that functional analysis of topoisomerase function and/or cleavable complexes was not done, which could further help to determine the effectiveness of these drugs. However, it is clear that fine-needle aspiration biopsy represents a easy means to access and evaluate a population of a given patient's tumor cells, which can be useful for biochemical and histologic analyses. We are currently developing an assay to determine the frequency of cleavable complexes in fine-needle aspiration specimens that may be useful in future trials.

Based on these results, we believe the topoisomerase inhibitor, karenitecin, has moderate activity in the current schedule in melanoma. We are currently examining this drug in oral form using a prolonged dosing in combination with another currently used antimelanoma agent to determine if higher response rates may be generated.

Footnotes

  • Grant support: NIH grant CA 82533.

  • 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 December 23, 2004.
    • Received August 25, 2004.
    • Revision received December 10, 2004.

References

  1. ↵
    Serrone L, Zeuli M, Sega FM, Cognetti F. Dacarbazine-based chemotherapy for metastatic melanoma: thirty-year experience overview. J Exp Clin Cancer Res 2000;19:21–34.
    OpenUrlPubMed
  2. ↵
    Wolchok JD, Klimek VM, Williams L, Chapman PB. Prophylactic recombinant epoetin α markedly reduces the need for blood transfusion in patients with metastatic melanoma treated with biochemotherapy. Cytokines Cell Mol Ther 1999;5:205–6.
    OpenUrlPubMed
  3. ↵
    Klimek VM, Wolchok JD, Chapman PB, Houghton AN, Hwu WJ. Systemic chemotherapy. Clin Plast Surg 2000;27:451–61, ix–x.
  4. ↵
    Atkins MB, Lee S, Flaherty L, Sosman JS, Sondak V, Kirkwood JM. A prospective randomized phase III trial of concurrent biochemotherapy (BCT) with cisplatin, vinblastine, dacarbazine (CVD), IL-2 and interferon α-2b (IFN) versus CVD alone in patients with metastatic melanoma (E3695): an ECOG-coordinated intergroup study Proc Am Soc Clin Oncol 2003;22:708 Abstract 2847.
    OpenUrl
  5. ↵
    Saltz LB, Pizzolato JF. The camptothecins. Lancet 2003;361:2235–42.
    OpenUrlCrossRefPubMed
  6. ↵
    Garcia-Carbonero R, Supko JG. Current perspectives on the clinical experience, pharmacology, and continued development of the camptothecins. Clin Cancer Res 2002;8:641–61.
    OpenUrlAbstract/FREE Full Text
  7. ↵
    Van Hattum A. New highly lipophilic camptothecin BNP 1350 is an effective drug in experimental human cancer. Int J Cancer 2000;88:260–6.
    OpenUrlCrossRefPubMed
  8. ↵
    Schilsky RL, Hausheer FH, Bertucci D, Berghorn E, Kindler HL, Ratain M. Phase I trial of karenitecin (KT) administered intravenously daily for five consecutive days in patients with advanced solid tumors using accelerated dose titration. Proc Am Soc Clin Oncol 2000;19:195.
    OpenUrl
  9. ↵
    Valkov NI, Gump JL, Sullivan DM. Quantitative immunofluorescence and immunoelectron microscopy of the topoisomerase IIα associated with nuclear matrices from wild-type and drug-resistant chinese hamster ovary cell lines. J Cell Biochem 1997;67:112–30.
    OpenUrlCrossRefPubMed
  10. ↵
    Whitacre CM, Zborowska E, Gordon NH, Mackay W, Berger NA. Topotecan increases topoisomerase IIα levels and sensitivity to treatment with etoposide in schedule-dependent process. Cancer Res 1997;57:1425–8.
    OpenUrlAbstract/FREE Full Text
  11. ↵
    Huncharek M, Caubet JF, McGarry R. Single-agent DTIC versus combination chemotherapy with or without immunotherapy in metastatic melanoma: a meta-analysis of 3273 patients from 20 randomized trials. Melanoma Res 2001;11:75–81.
    OpenUrlCrossRefPubMed
  12. ↵
    Khayat D, Coeffic D. Chemotherapy and chemoimmunotherapy for metastatic malignant melanoma: review and recent advances. Gan To Kagaku Ryoho 2000;27:238–47.
  13. ↵
    Chapman PB, Einhorn LH, Meyers ML, et al. Phase III multicenter randomized trial of the Dartmouth regimen versus dacarbazine in patients with metastatic melanoma. J Clin Oncol 1999;17:2745–51.
    OpenUrlAbstract/FREE Full Text
  14. ↵
    Janik JE, Miller LL, Korn EL, et al. A prospective randomized phase II trial of GM-CSF priming to prevent topotecan-induced neutropenia in chemotherapy-naive patients with malignant melanoma or renal cell carcinoma. Blood 2001;97:1942–6.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    Kraut EH, Walker MJ, Staubus A, Gochnour D, Balcerzak SP. Phase II trial of topotecan in malignant melanoma. Cancer Invest 1997;15:318–20.
    OpenUrlPubMed
  16. ↵
    Rosenthal MA, Green M, Wiernik P, Borden EC, Marsh JC, Haller DG. Epirubicin has modest single-agent activity in head and neck cancer but limited activity in metastatic melanoma and colorectal cancer: phase II studies by the Eastern Cooperative Oncology Group. Am J Clin Oncol 1998;21:509–12.
    OpenUrlCrossRefPubMed
  17. Lopez M, Perno CF, Papaldo P, Di Lauro L, Ganzina F, Barduagni A. Phase II study of epirubicin in advanced malignant melanoma. Invest New Drugs 1984;2:315–7.
    OpenUrlPubMed
  18. ↵
    Hurteloup P, Cappelaere P, Armand JP, Mathe G. Phase II clinical evaluation of 4′-epi-doxorubicin. Cancer Treat Rev 1983;67:337–41.
    OpenUrl
  19. ↵
    Maliepaard M, van Gastelen MA, Tohgo A, et al. Circumvention of breast cancer resistance protein (BCRP)-mediated resistance to camptothecins in vitro using non-substrate drugs or the BCRP inhibitor GF120918. Clin Cancer Res 2001;7:935–41.
    OpenUrlAbstract/FREE Full Text
  20. ↵
    Hausheer FH, Haridas K, Zhao M, et al. A novel class of orally active highly lipophilic topoisomerase I inhibitors. Proc Am Assoc Cancer Res 1998;39:420.
  21. ↵
    Dingemans AM, Witlox MA, Stallaert RA, van der Valk P, Postmus PE, Giaccone G. Expression of DNA topoisomerase IIα and topoisomerase IIβ genes predicts survival and response to chemotherapy in patients with small cell lung cancer. Clin Cancer Res 1999;5:2048–58.
    OpenUrlAbstract/FREE Full Text
  22. Costa MJ, Hansen CL, Holden JA, Guinee D Jr. Topoisomerase IIα: prognostic predictor and cell cycle marker in surface epithelial neoplasms of the ovary and peritoneum. Int J Gynecol Pathol 2000;19:248–57.
    OpenUrlCrossRefPubMed
  23. ↵
    Bredel M, Pollack IF, Hamilton RL, Birner P, Hainfellner JA, Zentner J. DNA topoisomerase IIα predicts progression-free and overall survival in pediatric malignant non-brainstem gliomas. Int J Cancer 2002;99:817–20.
    OpenUrlCrossRefPubMed
  24. ↵
    Jarvinen TA, Holli K, Kuukasjarvi T, Isola JJ. Predictive value of topoisomerase IIα and other prognostic factors for epirubicin chemotherapy in advanced breast cancer. Br J Cancer 1998;77:2267–73.
    OpenUrlPubMed
  25. ↵
    Dingemans AC, van Ark-Otte J, Span S, et al. Topoisomerase IIα and other drug resistance markers in advanced non-small cell lung cancer. Lung Cancer 2001;32:117–28.
    OpenUrlCrossRefPubMed
  26. ↵
    Mirski SE, Sparks KE, Yu Q, et al. A truncated cytoplasmic topoisomerase IIα in a drug-resistant lung cancer cell line is encoded by a TOP2A allele with a partial deletion of exon 34. Int J Cancer 2000;85:534–9.
    OpenUrlCrossRefPubMed
  27. Valkov NI, Sullivan DM. Tumor p53 status and response to topoisomerase II inhibitors. Drug Resist Updat 2003;6:27–39.
    OpenUrlCrossRefPubMed
  28. Ernst AI, Soltermann A, Sigrist JA, Widmer L, Gasser SM, Stahel RA. Ectopic expression of human topoisomerase IIα fragments and etoposide resistance in mammalian cells. Int J Cancer 2000;88:99–107.
    OpenUrlCrossRefPubMed
  29. ↵
    Valkov NI, Sullivan DM. Drug resistance to DNA topoisomerase I and II inhibitors in human leukemia, lymphoma, and multiple myeloma. Semin Hematol 1997;34:48–62.
    OpenUrlPubMed
  30. ↵
    Beidler DR, Cheng YC. Camptothecin induction of a time- and concentration-dependent decrease of topoisomerase I and its implication in camptothecin activity. Mol Pharmacol 1995;47:907–14.
    OpenUrlAbstract
  31. ↵
    Desai SD, Li TK, Rodriguez-Bauman A, Rubin EH, Liu LF. Ubiquitin/26S proteasome-mediated degradation of topoisomerase I as a resistance mechanism to camptothecin in tumor cells. Cancer Res 2001;61:5926–32.
    OpenUrlAbstract/FREE Full Text
PreviousNext
Back to top
Clinical Cancer Research: 11 (8)
April 2005
Volume 11, Issue 8
  • 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 Clinical Cancer Research 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.
Phase II Trial of Karenitecin in Patients with Malignant Melanoma: Clinical and Translational Study
(Your Name) has forwarded a page to you from Clinical Cancer Research
(Your Name) thought you would be interested in this article in Clinical Cancer Research.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Phase II Trial of Karenitecin in Patients with Malignant Melanoma: Clinical and Translational Study
Adil Daud, Nikola Valkov, Barbara Centeno, Jennifer Derderian, Patricia Sullivan, Pamela Munster, Patricia Urbas, Ronald C. DeConti, Elmer Berghorn, Zhenmei Liu, Frederick Hausheer and Daniel Sullivan
Clin Cancer Res April 15 2005 (11) (8) 3009-3016; DOI: 10.1158/1078-0432.CCR-04-1722

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Phase II Trial of Karenitecin in Patients with Malignant Melanoma: Clinical and Translational Study
Adil Daud, Nikola Valkov, Barbara Centeno, Jennifer Derderian, Patricia Sullivan, Pamela Munster, Patricia Urbas, Ronald C. DeConti, Elmer Berghorn, Zhenmei Liu, Frederick Hausheer and Daniel Sullivan
Clin Cancer Res April 15 2005 (11) (8) 3009-3016; DOI: 10.1158/1078-0432.CCR-04-1722
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
    • Patients and Methods
    • Results and Analyses
    • Discussion
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • Biomarker Analysis from the BERIL-1 Study
  • Radiation and TGFβ Blockade in Metastatic Breast Cancer
  • R-BEAM or 90YIT-R-BEAM for DLBCL
Show more Cancer Therapy: Clinical
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • CCR Focus Archive
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About Clinical Cancer Research

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

Copyright © 2021 by the American Association for Cancer Research.

Clinical Cancer Research
eISSN: 1557-3265
ISSN: 1078-0432

Advertisement