New Anticancer Agents in Clinical Development

Publication
Article
OncologyONCOLOGY Vol 9 No 11
Volume 9
Issue 11

A better understanding of the biology and biochemistry of the cancer cell has led to the development of various promising new antineoplastic compounds that are now undergoing phase I, II, and III clinical testing. These drugs include topoisomerase I inhibitors, such as camptothecin and its analogs 9-aminocamptothecin, irinotecan, and topotecan; the paclitaxel analog docetaxel; gemcitabine, an antimetabolite structurally related to cytarabine; and fluorouracil prodrugs and other thymidylate synthase (TS) inhibitors.

A better understanding of the biology and biochemistry of the cancer cell has led to the development of various promising new antineoplastic compounds that are now undergoing phase I, II, and III clinical testing. These drugs include topoisomerase I inhibitors, such as camptothecin and its analogs 9-aminocamptothecin, irinotecan, and topotecan; the paclitaxel analog docetaxel; gemcitabine, an antimetabolite structurally related to cytarabine; and fluorouracil prodrugs and other thymidylate synthase (TS) inhibitors. Another exciting approach to cancer treatment is the use of agents that induce a less malignant state by altering cellular phenotype. Such agents include angiogenesis inhibitors, differentiating agents, signal transduction inhibitors, and gene therapy.

Introduction

The 1990s is an exciting decade for oncologists. Intensive research and development programs during the 1980s and 1990s have resulted in new anticancer agents with unique mechanisms of action and significant clinical activity. Recently, three such agents were approved by the FDA: paclitaxel (Taxol), all-trans-retinoic acid, and vinorelbine (Navelbine). These agents have shown significant clinical activity in patients with refractory tumors, such as non-small-cell lung cancer, platinum-refractory ovarian cancer, and anthracycline-refractory breast cancer.

This article will review other promising compounds currently in clinical development. These drugs, which include topoisomerase I inhibitors, docetaxel (Taxotere), gemcitabine (Gemzar), and thymidylate synthase (TS) inhibitors, have significant preclinical activity and are now undergoing phase I, II, and III clinical testing. The hope is that these novel compounds represent the first of a long line of new agents developed as a result of our better understanding of the biology and biochemistry of the cancer cell.

Topoisomerase I Inhibitors

Topoisomerase I inhibitors are an exciting new class of antineoplastic agents currently undergoing clinical testing. These compounds are structurally related to camptothecin, a natural product isolated from the Chinese plant Camptothecin accuminata [1].

Topoisomerase I is a cellular enzyme involved in maintaining the topographic structure of DNA during translation, transcription, and mitosis [2]. The double helix structure of DNA creates torsional strain in a cell that must be overcome in order for replication and translation to proceed. DNA topoisomerases control and modify the topological state of DNA by creating a transient break in a single strand (topoisomerase I) or both complementary strands (topoisomerase II) of the DNA backbone [3]. These enzymes are capable of catalyzing many types of interconversions between DNA topological isomers. Examples of interconversions include catenation (interlocking of DNA circles) and decatenation, and knotting (passing one double strand of DNA through another strand) and unknotting [3].

It is now established that transient breakage of the DNA backbone by topoisomerases is accompanied by the formation of a covalent enzyme-DNA intermediate called the cleavable complex [4]. Inhibition of topoisomerase I by camptothecin and its analogs is accomplished by stabilization of the enzyme-DNA cleavable complex. This occurs after the cleavage step and causes the DNA and topoisomerase to be trapped in the cleavable complex. When camptothecin is removed, the DNA is reannealed (ie, the DNA backbone is resealed), and replication can proceed. Thus, inhibition of topoisomerase I blocks cellular RNA and DNA synthesis. The mechanism by which topoisomerase I inhibitors cause cell death is presently unknown [5].

Camptothecin

During extensive screening of random plant products by the Cancer Chemotherapy National Service Center in the late 1950s, a crude extract of C accuminata was found to have anticancer activity [1]. In 1966, Wall and coworkers [1] isolated this extract, camptothecin (Figure 1), which demonstrated significant anticancer activity in L1210 leukemia and Walker 256 carcinosarcoma [6,7]. In preclinical studies, hemorrhagic enterocolitis was the major dose-limiting toxicity [8].

Phase I and II Trials--In the late 1960s and early 1970s, camptothecin sodium underwent phase I and phase II testing. Phase I studies were performed using various dosing schedules: single-dose [8], daily [9], weekly [9], and daily for 5 days [10]. Although 5 of 18 patients demonstrated objective tumor responses to the drug in one phase I trial, phase II studies in patients with melanoma [11] and adenocarcinoma of the colon [12] were limited by severe hemorrhagic cystitis and unpredictable myelosuppression. As a result, further clinical development of camptothecin sodium was halted.

Development of Analogs--It wasn't until the 1980s, when inhibition of topoisomerase I was identified as the mechanism of action of camptothecin, that interest in this class of compounds was rekindled. In addition, it was found that the lactone ring (E-ring, which is pH labile) was critical to the activity of camptothecin, and thus the sodium salt used in earlier trials (which mainly comprised the carboxylate [inactive] form) might have been the reason for the lack of antitumor activity observed [13].

Structure-activity studies [14] revealed that modification of the A-ring improved water solubility and reduced protein binding. Therefore, analogs of camptothecin with increased water solubility and decreased protein binding were developed, with the anticipation that such modifications would enhance activity while decreasing the hemorrhagic cystitis and unpredictable myelosuppression. Currently, camptothecin and four of its analogs are in clinical development: 9-aminocamptothecin (Figure 1), GI147211, irinotecan (CPT-11), and topotecan.

Oral Camptothecin--Camptothecin is undergoing evaluation as an oral preparation. Giovanella and Natelson reported the preliminary results of a trial with oral camptothecin in which the dose-limiting toxicity was gastrointestinal [15]. In the 52 patients treated, 5 partial responses and 1 complete response were noted.

9-Aminocamptothecin

9-Amino-20(S)-camptothecin (9-AC) has demonstrated significant preclinical activity. In studies conducted at the National Cancer Institute that measured DNA strand breaks and cytotoxicity against HT-29 cell lines, 9-AC was found to be slightly more potent than topotecan and significantly more potent than CPT-11, but slightly less potent than SN-38 (the active metabolite of CPT-11) and camptothecin.

Clinical development of 9-AC has proceeded slowly due to its relative water insolubility. In a phase I study of 9-AC administered as a 72-hour continuous infusion in patients with solid tumors, dose-limiting neutropenia occurred at 59 mcg/m²/h [16]. Other toxicities (all grade 2) included nausea, vomiting, mucositis, and diarrhea. Further dose escalation in combination with granulocyte colony-stimulating factor (G-CSF, filgrastim; Neupogen) is currently under study.

GI147211

GI147211 is a new water-soluble analog of camptothecin. In human tumor xenograft models HT-29 and SW-48 (colon), PC-3 (prostate), and MX-1 (breast), GI147211 was 1.5 to 1.8 times more active than topotecan in suppressing growth. GI147211 has also been found to be 2.3 to 4.3 times more potent in inhibiting topoisomerase I activity than topotecan [17].

Based on its preclinical activity, GI147211 has recently undergone clinical testing with two dosing schedules: daily doses for 5 days and a 72-hour continuous infusion every 21 days. Reversible grade 3 and 4 neutropenia and thrombocytopenia have been observed with both schedules [18]. Phase II trials are underway using the daily for 5 days schedule.

Irinotecan

The initial preclinical and clinical development of irinotecan (CPT-11) was conducted primarily in Japan. In preclinical testing, irinotecan was found to be active against a broad spectrum of tumor models [19]. However, the decarboxylated metabolite SN-38 (7-ethyl-10-hydroxy-camptothecin; Figure 1) plays a major role in the antitumor activity of irinotecan in vivo [20].

The maximum tolerated dose (MTD) of irinotecan depends on the dose and schedule, with diarrhea and neutropenia being the major toxicities. Schedules employing a daily dosing schedule have demonstrated more neutropenia, whereas intermittent schedules have been associated with significant diarrhea [21]. The dose intensity on all the schedules has been approximately 100 mg/m²/wk [21]. However, in a recently published study from France [22], escalation of the irinotecan dose was accomplished by means of aggressive treatment of the diarrhea with antimotility agents. An MTD of 600 mg/m² given over 90 minutes every 3 weeks was reported, with neutropenia being dose-limiting.

Diarrhea--Irinotecan has been associated with two forms of diarrhea. The first type occurs during or just after the infusion and has a cholinergic mecha nism. The use of atropine at the onset of this early diarrhea is an effective treatment.

The second type of diarrhea begins 3 to 5 days after the irinotecan infusion and may be moderate to severe in 20% of patients. Aggressive treatment at the onset with antimotility agents may obviate its severity. If late diarrhea is not treated early, it usually runs a 5- to 7-day course. The mechanism of this type of diarrhea is unknown, but it may be secondary to the biliary excretion of SN-38, the active metabolite of irinotecan [23].

Other Toxicities--In addition to dose-limiting myelosuppression and diarrhea, other toxicities reported with irinotecan include anemia, transaminasemia, anorexia, alopecia, malaise, flushing, stomatitis, pneumonitis, nausea, and vomiting. These toxicities are mild to moderate in severity and reversible [21].

Antitumor Activity--Single-agent activity of irinotecan has been evaluated in a number of tumor types, including non-Hodgkin's and Hodgkin's lymphoma, acute leukemia, colon cancer, non-small-cell and small-cell lung cancer, ovarian cancer, cervical cancer, breast cancer, pancreatic cancer, and gastric cancer (Table 1). The encouraging activity of this agent seen in patients with refractory tumors, such as cervical cancer and colon cancer, has stimulated large phase II trials now being conducted in the United States and abroad.

Irinotecan Combinations--Because of the novel mechanism of action and clinical activity of irinotecan, investigators have explored its use in combination with other cytotoxic agents. In vitro and in vivo testing of camptothecin analogs has demonstrated synergistic activity when combined with topoisomerase II inhibitors, alkylating agents, platinum compounds, and radiation [24].

Phase I trials of several irinotecan combinations have been initiated. Impressive activity has been demonstrated when irinotecan is combined with cisplatin (Platinol) or etoposide (VePesid) in patients with non-small-cell lung cancer. In untreated patients with non-small-cell lung cancer, response rates to irinotecan-cisplatin have ranged from 43% to 45% [44,45]. These results have prompted further investigation of this combination.

Topotecan

Topotecan is a semisynthetic analog of camptothecin that incorporates a stable basic side chain at the 9-position of the A-ring of 10-hydroxycamptothecin (Figure 1). This basic side chain affords water solubility without requiring hydrolysis of the E-ring lactone.

Based on the preclinical activity seen with topotecan, several phase I clinical studies were initiated. To date, 18 phase I studies using nine different schedules of topotecan have been reported [21].

Cytopenias--The dose-limiting toxicity observed with all schedules of topotecan (except in leukemia, for which mucositis was dose-limiting) has consisted of neutropenia and thrombocytopenia. Neutropenia and mild thrombo- cytopenia were seen with the short infusion schedules, whereas schedules with longer infusion times resulted in both dose-limiting neutropenia and thrombocytopenia. The neutropenia and thrombocytopenia have generally been short-lived (less than 7 days) and rarely associated with fever.

Other toxicities observed have included mild nausea and vomiting, anorexia, diarrhea, alopecia, fatigue, and skin rash. Unlike camptothecin, hemorrhagic cystitis has not been observed with topotecan, and unlike irinotecan, diarrhea has not been a significant problem.

Efforts at Dose Escalation--Because the major toxicities of topotecan are hematologic, the use of G-CSF to permit further dose escalation of this camptothecin analog has been explored. Unfortunately, no significant dose escalation has been possible with colony-stimulating factors as a result of the development of dose-limiting thrombocytopenia [47].

Use in Hepatic or Renal Dysfunction--Topotecan also has been evaluated in patients with hepatic or renal dysfunction. Patients with renal insufficiency had significantly decreased topotecan clearance requiring dose reduction. Patients with hepatic dysfunction did not exhibit altered drug clearance and were able to tolerate doses similar to those used in patients with normal hepatic function [48].

Antitumor Activity--The phase II activity of topotecan is summarized in Table 2. Encouraging activity in patients with previously treated small-cell-lung cancer and ovarian cancer has stimulated phase III trials in these patient populations. Like irinotecan, topotecan has been combined with various antineoplastic agents and with radiotherapy. Phase I studies of the combination of topotecan and etoposide, doxorubicin, cisplatin, paclitaxel, and radiation have been reported (Table 3).

Oral Preparation--Topotecan has recently undergone testing as an oral preparation. The oral route has the potential advantage of ease of administration. In addition, the acidic pH of the stomach should maintain topotecan in the active closed lactone form. Creemers et al reported their experience with topotecan given orally on day one followed by an intravenous dose on day two [66]. They found that the oral form exhibited 32% bioavailability and was not affected by first-pass metabolism.

Docetaxel

Docetaxel (N-debenzoyl-N-tert-butoxycarbonyl-10-deacytyl taxol, RP56976 [Taxotere]) is a semisynthetic analog of paclitaxel prepared from a noncytotoxic precursor extracted from the needles of the European yew tree Taxus baccata (Figure 2). Docetaxel was synthesized in 1986 and was selected for clinical development in 1987 due to its preclinical activity and a formulation that allowed for shorter infusion schedules than paclitaxel.

Early preclinical testing demonstrated that docetaxel is 2.5-fold more potent than paclitaxel [67]. Like paclitaxel, docetaxel binds to tubulin, promotes the assembly of microtubules, and inhibits depolymerization [67].

Phase I Testing

In 1990, phase I testing of taxotere began in Europe and the United States. Six dosing schedules have been explored, with neutropenia being the dose-limiting toxicity on all schedules [67]. Studies that have utilized longer schedules or repeated-dose schedules have found both mucositis and neutropenia to be dose-limiting. Mucositis was not found to be a significant problem on 1- and 2-hour dosing schedules.

Hematologic Toxicity--The neutropenia associated with docetaxel is dose-dependent, with a median time to neutrophil nadir of 9 days and recovery by day 21. At the recommended phase II dose, approximately 70% of patients develop grade 4 neutropenia; of these patients, only 18% have grade 4 neutropenia lasting for more than 7 days, and 15% have fever associated with the neutropenia. Anemia and thrombocytopenia are generally mild (grade 2) and seen only in a minority of patients (20%).

Hypersensitivity--In the phase I studies, none of the patients received premedication for hypersensitivity to docetaxel. In 15% of patients, a reaction characterized by localized or generalized flushing, rash, chest pain or heaviness, back pain, dyspnea, and fever was observed [68]. Such reactions occurred within 3 to 10 minutes after the infusion was begun and resolved within a few minutes after it was interrupted. Treatment with diphenhydramine and hydrocortisone allowed the infusion to be restarted.

Severe symptoms associated with hypotension and/or bradycardia were seen in only 2% of patients. Premedication with antihistamines and steroids reduced the incidence of the hypersensitivity reaction [69]. Patients who developed the hypersensitivity reaction did so in the first or second cycle of therapy [68].

Other toxicities reported for docetaxel have been generally mild (grade 1 and 2) and include alopecia (100% of patients), mucositis (49% on the longer infusion schedules), skin reactions (69%), edema (40%), weakness (27%), and neuropathy (31%) [70].

The edema has been associated with the development of pleural effusions and ascites. The etiology of this fluid collection syndrome is unknown but may be related to increased capillary permeability [71]. Edema appears to occur after a total dose of approximately 400 to 500 mg/m² and can be delayed by premedication with steroids, antihistamines, and the use of diuretics.

The neuropathy is a sensory neuropathy that is generally mild (grade 1 or 2), with symptoms of numbness and dysesthesias. It resolves slowly when docetaxel is discontinued [72].

Pharmacokinetics

Population pharmacokinetics of docetaxel have been determined. Bruno et al [73], using a three-compartment model, found docetaxel to have a half-life of 11.1 hours with a clearance of 21.2 L/h/m² and a volume of distribution of 69 L/m². Metabolism studies have demonstrated that 80% of docetaxel is recovered in the feces over 7 days, with only 5% recovered in the urine [74].

Antitumor Activity

In view of the activity noted in phase I trials, docetaxel underwent broad phase II testing (Table 4a and Table 4b ). Clinical activity has been documented in patients with a number of tumor types, including breast cancer, non-small-cell lung cancer, ovarian cancer, head and neck cancer, soft-tissue sarcomas, and gastric cancer. The major activity of docetaxel observed in the treatment of patients with breast cancer and non-small-cell lung cancer has sparked intense interest.

Breast Cancer--Docetaxel has dem onstrated objective responses in both untreated and treated patients with metastatic breast cancer, with response rates reported to be between 44% and 58% in both of these groups [75-79]. This activity is comparable to the most active single agents in breast cancer, including doxorubicin and paclitaxel.

Non-Small-Cell Lung Cancer--In non-small-cell lung cancer, responses to docetaxel have been documented in both chemotherapy-naive patients and in those whose disease has progressed after a platinum-containing regimen. Published response rates range from 21% to 33% [80-85]. This activity in platinum-refractory patients is notable when compared to that of other agents in this population. For example, paclitaxel, cisplatin, etoposide, epidoxorubicin (Epirubicin), and vindesine (Eldisine) all have published response rates ranging from 2% to 10% when used as second-line treatment.

Ongoing Studies

Various studies of docetaxel are currently underway. These include: phase I studies of the combination of docetaxel with cisplatin and with fluorouracil; phase II trials in paclitaxel-resistant breast cancer, hormone-refractory prostate cancer, and cholangiocarcinoma; a phase III trial of docetaxel vs best supportive care as second-line treatment for patients with non-small-cell lung cancer; and a phase III trial of docetaxel vs paclitaxel in anthracycline-refractory breast cancer.

Gemcitabine

Gemcitabine (2'2'-difluorodeoxycytidine; dFdC [Gemzar]) is a synthetic pyrimidine antimetabolite structurally related to cytarabine. Gemcitabine differs from the endogenous nucleoside, deoxycytidine, by the presence of two fluorine atoms in its deoxyribofuranosyl ring (Figure 1).

Gemcitabine inhibits both RNA and DNA viruses in cell culture and was originally synthesized as an antiviral agent. Its narrow therapeutic index during in vivo evaluation, however, precluded further development as an antiviral drug [101]. Gemcitabine was subsequently found to have excellent in vitro antineoplastic activity in tumor cell lines, as well as broad-spectrum activity against a panel of murine solid tumors and human tumor xenografts [101].

Mechanism of Cytotoxicity

Gemcitabine's cytotoxic activity is due to the inhibition of DNA synthesis and repair. It is a prodrug requiring intracellular metabolic activation to its phosphorylated forms by deoxycytidine kinase. Gemcitabine triphosphate competes with deoxycytidine triphosphate (dCTP) as a substrate for incorporation into DNA. Once incorporated into DNA, gemcitabine triphosphate causes a profound inhibition of DNA elongation and chain termination. Gemcitabine diphosphate is an inhibitory substrate for ribonucleotide reductase (RbNR), an enzyme required for the production of deoxynucleotides used for DNA synthesis and repair. Cell death associated with these events exhibits the morphologic and biochemical characteristics of apoptosis.

Self-Potentiation

Gemcitabine appears to have the ability to enhance its own activity (self-potentiation). Inhibition of RbNR causes lowering of intracellular dCTP levels. Through feedback mechanisms, low dCTP levels activate deoxycytidine kinase and inactivate deoxycitidine monophosphate (dCMP) deaminase, leading to increased phosphorylation (activation) and decreased deamination (elimination) of gemcitabine. Low intracellular levels of dCTP also enhance gemcitabine triphosphate's incorporation into DNA, due to competition for DNA poly merase. These mechanisms may explain the increased cellular accumulation and increased activity of gemcitabine seen in solid tumors, as compared with cytarabine.

Phase I Trials

Toxicities of gemcitabine observed in phase I trials have included myelosuppression, reversible skin rash, fever, mild nausea and vomiting, alopecia, lethargy, a flu-like syndrome, peripheral edema, and reversible elevations in liver function tests. Cumulative toxicity has not been observed.

In phase I trials, dose-limiting toxicity of gemcitabine has been found to be particularly schedule dependent; ie, there is a marked difference in the minimum tolerated dose (MTD) depending on the schedule of administration. Dose-limiting lethargy and flu-like syndrome were seen on the daily doses for 5 days schedule, whereas myelosuppression was dose-limiting on the every other week schedule.

Antitumor Activity

Clinical activity was demonstrated in patients with non-small-cell lung, pancreatic, breast, head and neck, bladder, renal cell, and colon cancer. Among the dosing schedules studied (daily doses for 5 days, twice a week, weekly, and biweekly), the weekly schedule was chosen for phase II trials due to adequate dose intensity and preclinical data suggesting schedule-related efficacy [102].

Based on the activity profile seen in phase I trials, broad phase II testing with gemcitabine is being conducted in a wide variety of malignancies. As summarized in Table 1, activity has been confirmed in patients with non-small-cell lung and small-cell lung cancers, breast cancer, ovarian cancer, pancreatic cancer, and squamous cell carcinoma of the head and neck.

Pharmacokinetics

The pharmacokinetics of gemcitabine have been extensively studied. Gemcitabine undergoes intracellular phosphorylation by deoxycytidine kinase to its active metabolites. Deamination to its uridine metabolites (dFdU[difluorodeoxy-uridine]) by cytidine deaminase is the principal mechanism involved in the elimination of gemcitabine.

Following a 30-minute intravenous infusion, gemcitabine has a short terminal half-life ranging from 4 to 20 minutes [121]. The deaminated metabolite (dFdU), however, has a much longer half-life (4 to 24 hours). Gemcitabine clearance is lower in women than in men, perhaps related to differences in the activity of cytidine deaminase [122].

Ongoing Trials

A determination of gemcitabine's clinical benefit, as assessed by changes in pain, performance status, and weight, is the primary objective of two ongoing clinical trials in patients with advanced pancreatic cancer [123].

Gemcitabine's favorable toxicity profile makes it an attractive candidate for combination therapy with other antineoplastic agents. Several trials utilizing gemcitabine combined with other agents, such as cisplatin (Platinol), carboplatin (Paraplatin), paclitaxel (Taxol), and hydroxyurea (Hydrea) are currently underway.

TS Inhibitors and Fluorouracil

Prodrugs

Fluorouracil was the first agent specifically synthesized to exploit an observed metabolic difference between normal and tumor cells [124]. It has been used either alone or in combination with other antineoplastic drugs for the treatment of a wide variety of tumors.

Fluorouracil is metabolized through different biochemical pathways, producing different cytotoxic metabolites, and is eliminated through urinary excretion (15% to 20%) or by the liver and extrahepatic tissues (80%). Three different mechanisms of action are thought to be responsible for its cytotoxicity:

  • Inhibition of the enzyme thymidylate synthase (TS) by its metabolite fluorodeoxyuridine monophosphate (FdUMP);
  • Incorporation of another metabolite, fluorouridine triphosphate (FUTP), into cellular RNA; and
  • Incorporation of fluorodeoxyuridine triphosphate (FdUTP) into cellular DNA (Figure 2).

Modulation of Fluorouracil Metabolism-Enhancement of the action of fluorouracil through modulation of its metabolic pathways has been pursued avidly for several years. The best known of these modulations is achieved by the use of 5-formyl-tetrahydrofolate (leucovorin), which stabilizes the TS-FdUMP-folate complex (Figure 2).

Other drugs have also been shown to enhance the cytotoxicity of fluorouracil. Dipyridamole inhibits the uptake of thymidine, decreasing the salvage pathway to overcome FdUMP inhibition of TS. Phosphonacetyl-L-aspartate (PALA) decreases intracellular uridine triphosphate and increases incorporation of FUTP into RNA. Methotrexate, when given before fluorouracil, increases intracellular phosphoribosyl pyrophosphate (PRPP), a substrate necessary for the enzyme-dependent conversion of fluorouracil to fluorouridine monophosphate (FUMP) (Figure 2). Interferons increase the conversion of fluorouracil to FdUMP, reduce levels of TS, and inhibit thymidine salvage pathways. Hydroxyurea, through inhibition of RbNR, depletes endogenous levels of deoxyuridine monophosphate (DUMP) and impairs FdUMP binding to TS (Figure 2). Thymidine and uracil have also been explored as fluorouracil-enhancing agents; these agents inhibit fluorouracil catabolism.

Given the multiple targets of fluorouracil and its complicated metabolism, attempts to create a more efficient drug with similar targets have been the focus of research over the last few years.

Fluorouracil Prodrugs

Several analogs of fluorouracil have been tested in clinical trials. These include 5-fluoro-2¢-deoxyuridine (floxuridine, FdUrd [FUDR]) and 5-deoxy-5-fluorouridine (dFUrd). Rapid degradation of floxuridine and the significant neurotoxicity produced by dFUrd have limited their use.

Tegafur (Ftorafur) is fluorouracil linked to a furan-ring dehydroxylated ribose sugar. Hydroxylation of tegafur by hepatic microsomal enzymes releases fluorouracil, leading to a slow but sustained level of fluorouracil in tumor cells.

Tegafur has exhibited considerable central nervous system (CNS) and gastrointestinal toxicity with no significant clinical benefits when compared to fluorouracil [125]. These findings halted studies in the United States; however, studies abroad have shown that fractionation of the daily dose, when given either intravenously or orally, produces considerably fewer side effects and improved clinical benefit [126].

Neurotoxicity has been a significant obstacle to the development of fluorouracil prodrugs and related oral compounds. Histopathologic studies in necropsies of humans and animals that received these drugs have shown vacuolization and necrosis/softening-like changes in the CNS. These alterations are thought to be produced by two fluorouracil catabolites, monofluoroacetic acid and, especially, alpha-fluoro-beta-alanine. The mechanism of this effect is not well understood [127].

Inhibitors of Fluorouracil Catabolism

Dihydropyrimidine dehydrogenase (DPd) is the initial and rate-limiting enzyme in the catabolism of fluorouracil, uracil, thymine, and other 5-substituted pyrimidines. More than 80% of administered fluorouracil is rapidly degraded by this enzyme. Fluorouracil half-life is approximately 5 to 20 minutes in individuals with normal levels of this enzyme, whereas persons with an inherited deficiency of DPd have a 10-fold longer half-life of the drug. Effective inhibition of this enzyme would theoretically increase the serum levels, half-life, and, possibly, efficacy of fluorouracil (Figure 2).

UFT--Two approaches have taken advantage of inhibition of this enzyme. The first approach, known as UFT, consists of an oral form of tegafur with uracil in a 1:4 molar concentration. Uracil inhibits the hepatic activity of DPd, thereby increasing the plasma levels of fluorouracil resulting from tegafur metabolism. The ease of administration of UFT and preliminary studies in Japan have stimulated interest in this drug. A phase II study showed response rates comparable to intravenous fluorouracil in patients with colorectal and gastric carcinomas [128].

Modulation of UFT with oral leucovorin has been attempted as a way to enhance its efficacy [129]. Pazdur et al recently reported a phase II trial using this combination in 45 colon cancer patients (42 of whom were previously untreated and 3 of whom had received adjuvant chemotherapy) [130]. Both drugs were given in three divided doses daily (300 to 350 mg/m² of UFT plus 150 mg/d of leucovorin) for 28 days, followed by a 7-day rest period. After two courses of therapy, 18 patients had a partial response and 1 patient had a complete response (42% response rate).

5-Ethynyluracil--The second approach is 5-ethynyluracil (776C85), which binds to and irreversibly inactivates DPd. This drug has been demonstrated to markedly increase the area under the curve, oral bioavailability, and therapeutic index of fluorouracil in animals [131]. Recent data in animals suggest that this compound is considerably more effective than high-dose uracil in sustaining plasma fluorouracil generated from tegafur [132]. In addition, 776C85 has been shown to protect against fluorouracil-induced neurotoxicity in dogs. This may occur through inhibition of the formation of neurotoxic catabolites or through the protection conferred by high serum levels of uracil [133].

Phase I trials testing 5-ethynyluracil in combination with fluorouracil, with or without leucovorin modulation, are currently underway in San Antonio and at the University of Chicago.

New TS Inhibitors

Thymidylate synthase catalyzes the conversion of DUMP by reductive methylation into deoxythymidine monophosphate (dTM) and dihydrofolate. This reaction requires the presence of a reduced folate cofactor and provides the precursor of deoxythymidine triphosphate (dTT), one of the deoxyribonucleotides necessary for DNA synthesis. Inhibition of this enzyme is probably the most important mechanism of action of fluorouracil and related compounds (Figure 2). Total TS activity, as measured by a catalytic assay (ie, the capacity to convert DUMP to dTM) and TS inhibition have been shown to correlate with response to fluorouracil [134]. An array of new agents has been developed to produce more effective inhibition of this enzyme.

CB3717 is a quinazoline-based TS inhibitor that competes with reduced folates, producing potent inhibition of the enzyme. Unfortunately, it has poor aqueous solubility and produces life-threatening nephrotoxicity and dose-independent hepatotoxicity.

Tomudex--As an alternative to CB3717, Tomudex (ZD1694, ICI D1694) was synthesized. Tomudex has the advantage of water solubility and rapid intracellular polyglutamation. Its polyglutamate derivatives are more active against TS and are retained longer within the cell, giving this compound more efficacy in vivo [135].

Phase I trials of Tomudex in Europe recommended a dose of 3.0 mg/m² given over 15 minutes every 21 days. Dose-limiting toxicities were diarrhea, neutropenia, malaise, and asthenia. Dose escalation of Tomudex up to 4.5 mg/m² has been reported [136], with a recommended phase II dose of 4 mg/m². These higher doses have produced increased myelosuppression and elevated hepatic transaminases.

Phase II trials of Tomudex using the 3-mg/m² dose have demonstrated significant activity in patients with advanced colorectal carcinoma (31/124 patients [25%], with 2 complete responses), breast cancer (11/45 patients [24%], with 2 complete responses), as well as in patients with pancreatic, non-small-cell lung cancer, and ovarian cancer [137]. Minimal toxicity was observed in these trials.

Phase III trials will randomize patients with colorectal cancer to either Tomudex or a standard 5-day regimen of fluorouracil/leucovorin.

AG-331--Other folate antimetabolites undergoing clinical trials are AG-331 and LY231514. AG-331 lacks a glutamate moiety but is very lipophilic, which facilitates tissue penetration. The lack of polyglutamation contributes to its favorable toxicity profile. Phase I trials with both 1- and 24-hour continuous infusions, daily for 5 days, are currently being conducted.

LY231514 undergoes extensive intracellular polyglutamation, which results in a more sustained drug effect. Data from two phase I studies are available [138]. In the first study, the drug was administered weekly for 4 weeks every 42 days. Dose-limiting neutropenia was encountered at 40 mg/m². Two minor responses in patients with colorectal cancer were documented.

In the second study, LY231514 was administered every 21 days. Reversible neutropenia was the dose-limiting toxicity, with dose escalation up to 600 mg/m². Minor responses in six patients with colorectal cancer and partial responses in two patients with pancreatic cancer were reported.

New Approaches To Anticancer Therapy

The chemotherapeutic compounds described above have direct cytotoxic effects on cancer cells through interaction with DNA, RNA, or protein synthesis. Another approach to cancer therapy is the use of agents that alter the cellular phenotype and thereby induce a less malignant state. Examples of such agents are angiogenesis inhibitors, differentiating agents, signal transduction inhibitors, and gene therapy.

This approach to treating cancer is quite novel, and the clinical development of these compounds is evolving. Standard cytotoxic agents produce tumor shrinkage and objective responses, whereas these agents may produce only cytostasis, and therefore, stable disease. Standard phase I and II clinical trials are not designed to assess end points such as time to tumor progression or time to relapse. A successful development strategy for these compounds with unique mechanisms of action must include early randomized trials in which the novel agent is/is not given in combination with a standard agent or is/is not given after adjuvant therapy. Randomized trials are more expensive, but are equipped to evaluate whether these novel agents should become part of a standard therapeutic regimen.

The section below will focus on three angiogenesis inhibitors that are currently undergoing phase I/II testing.

Angiogenesis Inhibitors

Abundant preclinical literature demonstrates that tumors are dependent on angiogenesis for tumor growth and metastasis [139]. In human breast cancer, angiogenesis is an independent negative prognostic factor in node-negative tumors [140]. These data support the significance of angiogenesis in tumor biology and suggest that it can be utilized as a target for novel therapeutic strategies. Angiogenesis inhibitors fall into two broad categories: protease inhibitors (which inhibit the proteases required for the penetration of the basement membrane by endothelial cells) and endothelial cell growth-factor inhibitors (which inhibit the growth factors required by endothelial cells).

Tecogalan sodium, recombinant human platelet-factor 4, and TNP-470 are three angiogenesis inhibitors that are currently being tested in cancer patients.

Tecogalan sodium is a sulfated polysaccharide polypeptidoglycan isolated from the cell walls of the bacterium Arthrobacter [141]. Its antiangiogenic effect is thought to be mediated by inhibition of the binding of basic fibro-
blast growth factor to endothelial cell receptors.

Phase I clinical trials of tecogalan in solid tumors and in AIDS-related Kaposi's sarcoma (KS) are currently being conducted at several sites. The primary toxicities observed to date have been fever, rigors, and prolongation of the activated partial thromboplastin and prothrombin times [142]. The coagulation toxicities have been ameliorated by prolonging the infusion duration at a given dose. Several standard dosing schedules of this compound are being investigated, as well as prolonged administration by continuous intravenous infusion.

Recombinant human platelet factor 4 is an antiangiogenic protein undergoing phase I/II testing in metastatic colon carcinoma and AIDS-related KS. When used via an intralesional injection in AIDS-related KS, this compound produced a 57% response rate in injected lesions (two complete responses) [143]. The primary toxicity was pain at the injection site. A dose-finding study in metastatic colon cancer examined schedules utilizing 30-minute infusions given up to 5 times a week; no significant biochemical, hematologic, or coagulation toxicities were noted [144].

TNP-470 is a fumigillin analog that has demonstrated potent antiangiogenic activity both in vitro and in vivo [145]. Phase I/II studies are currently being conducted in AIDS-related KS and hormone-refractory prostate cancer. Although no measurable responses have been reported, in one AIDS-related KS trial two patients given the higher dose levels experienced significant reductions in painful extremity edema [146]. Toxicities have been mild, and consist primarily of fatigue without muscle weakness [147].

Summary

The treatment of patients with advanced malignancies has often been discouraging, owing to the poor response rates and significant toxicity of currently available antineoplastic agents. Recently, various new agents with novel mechanisms of action have been developed and are in clinical trials. For the first time in decades, the possibility of improvement in the treatment of patients with advanced and refractory malignancies exists.

Encouraging phase I and II activity of camptothecin and its analogs has stimulated further development of these agents alone and in combination with other cytotoxic agents. Similarly, gemcitabine is being studied alone in patients with advanced pancreatic cancer, and as a component of combination regimens. The promising results obtained with docetaxel in patients with metastatic breast cancer, as well as in patients with non-small-cell lung cancer, also warrant further study.

Finally, an understanding of the metabolism and mechanisms of action of fluorouracil has created new possibilities to enhance its clinical activity. These new modulators extend the spectrum of activity of fluorouracil. Continued evaluation of these exciting compounds will further define their role in the management of cancer patients.

References:

1. Wall ME, Wani MC, Cook CE, et al: Plant anti-tumor agents. 1. The isolation and structure of camptothecin, a novel alkaloidal leukemia and tumor inhibitor from Camptotheca Acuminata. J Am Chem Soc 88(3):3888-3890, 1966.

2. Zijlstra JG, De Jong S, De Vries EG, et al: Topoisomerases, new targets in cancer chemotherapy. Med Oncol Tumor Pharmacother 7:11-18, 1990.

3. D'Arpa P, Liu LF: Topoisomerase-targeting antitumor drugs. Biochim Biophys Acta 989:163-177, 1989.

4. Wang JC: DNA Topoisomerases. Ann Rev Biochem 54:665-697, 1985.

5. Hsiang YH, Lihou MG, Liu LF: Arrest of replication forks by drug-stabilized topoisomerase I-DNA cleavable complexes as a mechanism of cell killing by camptothecin analogues. Cancer Res 49:5077-5082, 1989.

6. DeWys WD, Humphreys SR, Goldin A: Studies on therapeutic effectiveness of drugs with tumor weight and survival time indices of Walker 256 carcinosarcoma. Cancer Chemother Rep 52:229-242, 1968.

7. Venditti JM, Abbott BJ: Studies on oncolytic agents from natural sources. Correlations of activity against animal tumors and clinical effectiveness. Lloydia 30:332-348, 1967.

8. Gottlieb JA, Guarino AM, Call JB, et al: Preliminary pharmacologic and clinical evaluation of camptothecin sodium (NSC-100880). Cancer Chemother Rep 54(6):461-470, 1970.

9. Muggia FM, Creaven PJ, Hansen HH, et al: Phase I clinical trial of weekly and daily treatment with camptothecin (NSC-100880): Correlation with preclinical studies. Cancer Chemother Rep 56(4):515-521, 1972.

10. Creaven PJ, Allen LM, Muggia FM: Plasma camptothecin (NSC-100880) levels during a 5-day course of treatment: Relation to dose and toxicity. Cancer Chemother Rep 56(5):573-578, 1972.

11. Gottlieb JA, Luce JK: Treatment of malignant melanoma with camptothecin (NSC-100880). Cancer Chemother Rep 56(1):103-105, 1972.

12. Moertel CG, Schutt AJ, Reitemeier RJ, et al: Phase II study of camptothecin (NSC-100880) in the treatment of advanced gastrointestinal cancer. Cancer Chemother Rep 56(1):95-101, 1972.

13. Wall ME, Wani MC: Camptothecin. Anticancer Agents Based on Natural Product Models, in Cassidy JM (ed). Chapter 12, Academic Press, Inc, pp 417-436, 1980.

14. Jaxel C, Kohn KW, Wani MC, et al: Structure-activity study of the actions of camptothecin derivatives on mammalian topoisomerase: I. Evidence for a specific receptor site and a relation to antitumor activity. Cancer Res 49:1465-1469, 1989.

15. Giovanella BC, Natelson E: Preclinical and clinical trials of oral 20-(S)-camptothecin (CPT) and of 9-nitro-so-(S)-camptothecin (9NC) (abstract). Fifth Conference on DNA Topoisomerases in Therapy 31:A32, 1994.

16. Dahut W, Brillhart N, Takimoto C, et al: A phase I trial of 9-aminocamptothecin (9-AC) in adult patients with solid tumors (abstract). Proc Am Soc Clin Oncol 13:A345, 1994.

17. Emerson DL, Besterman JM, Brown HR et al: In vivo antitumor activity of two new seven-substituted water-soluble camptothecin analogues. Cancer Res 55(3): 603-609, 1995.

18. Wissel P, Verweij J, Eckardt J: On-going phase I trials on intravenous GI147211, a totally synthetic camptothecin analog, administered by the daily 5 and 72 hour CI regimens (abstract). Fifth Conference on DNA Topoisomerases in Therapy 32:A33, 1994.

19. Kunimoto T, Nitta K, Tanaka T, et al: Anti tumor activity of 7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxy-camptothecin, a novel water-soluble derivative of camptothecin, against murine tumors. Cancer Res 47:5944-5947, 1987.

20. Kaneda N, Nagata H, Furuta T, et al: Metabolism and pharmacokinetics of the camptothecin analogue CPT-11 in the mouse. Cancer Res 50:1715-1720, 1990.

21. Burris HA, Fields SM: Topoisomerase inhibitors: An overview of the camptothecin analogs. Hematol Onc Clin North Am 8(2):333-355, 1994.

22. Abigerges D, Chabot GG, Armand JP, et al: Phase I and pharmacokinetic studies of the camptothecin analog irinotecan administered every 3 weeks in cancer patients. J Clin Oncol 13(1):210-221, 1995.

23. Rowinsky EK, Grochow LB, Ettinger DS, et al: Phase I and pharmacological study of the novel topoisomerase I inhibitor 7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxy camptothecin (CPT-11) administered as a 90-minute infusion every 3 weeks. Cancer Res 54(2):427-436, 1994.

24. Kaufmann SH: Induction of endonucleolytic DNA cleavage in human acute myelogenous leukemia cells by etoposide, camptothecin, and other cytotoxic anticancer drugs: A cautionary note. Cancer Res 49:5870-5878, 1989.

25. Ohno R, Okada K, Masaoka T, et al: An early phase II study of CPT-11: A new derivative of camptothecin, for the treatment of leukemia and lymphoma. J Clin Oncol 8(11):1907-1912, 1990.

26. Shimada Y, Yoshino M, Wakui A, et al: Phase II study of CPT-11, new camptothecin derivative, in the patients with metastatic colorectal cancer (abstract). Proc Am Soc Clin Oncol 10:A408, 1991.

27. Fukuoka M, Negoro S, Niitani H, et al: Phase II study of a new camptothecin derivative, CPT-11 in previously untreated non-small-cell lung cancer (NSCLC) (abstract). Proc Am Soc Clin Oncol 9:A873, 1990.

28. Takeuchi S, Takamizawa H, Takeda Y, et al: Clinical study of CPT-11, camptothecin derivative, on gynecological malignancy (abstract). Proc Am Soc Clin Oncol 10:A617,1991.

29. Takeuchi S, Noda K, Yakushiji M: Late phase II study of CPT-11, a topoisomerase I inhibitor, in advanced cervical carcinoma (abstract). Proc Am Soc Clin Oncol 11:A708, 1992.

30. Negoro S, Fukuoka M, Niitani H, et al: Phase II study of CPT-11, new camptothecin derivative, in small cell lung cancer (SCLC) (abstract). Proc Am Soc Clin Oncol 10:A822, 1991.

31. Tsuda H, Takatsuki K, Ohno R, et al: A late phase II trial of a potent topoisomerase I inhibitor, CPT-11, in malignant lymphoma (abstract). Proc Am Soc Clin Oncol 11:A1070, 1992.

32. Ogawa M, Taguchi T: Clinical studies with CPT-11: the Japanese experience (abstract). Ann Oncol 3(suppl 1):118, 1992.

33. Fukuoka M, Niitani H, Suzuki A, et al: A phase II study of CPT-11, a new derivative of camptothecin, for previously untreated non-small-cell lung cancer. J Clin Oncol 10(1):16-20, 1992.

34. Masuda N, Fukuoka M, Kusunoki Y, et al: CPT-11: A new derivative of camptothecin for the treatment of refractory or relapsed small-cell lung cancer. J Clin Oncol 10(8):1225-1229, 1992.

35. Bonneterre J, Pion JM, Adenis A, et al: A phase II study of a new camptothecin analog CPT-11 in previously treated advanced breast cancer patients (abstract). Proc Am Soc Clin Oncol 12:A179, 1993.

36. Sakata Y, Wakui A, Nakao I, et al: A late phase II study of irinotecan (CPT-11), in advanced pancreatic cancer (abstract). Proc Am Soc Clin Oncol 12:A633, 1993.

37. Nakagawa K, Fukuoka M, Niitani H: Phase II study of irinotecan (CPT-11) and cisplatin in patients with advanced non-small-cell lung cancer (NSCLC) (abstract). Proc Am Soc Clin Oncol 12:A1104, 1993.

38. Shimada Y, Yoshino M, Wakui A, et al: Phase II study of CPT-11, a new camptothecin derivative, in metastatic colorectal cancer: CPT-11 Gastrointestinal Cancer Study Group. J Clin Oncol 11(5):909-913, 1993.

39. Rothenberg ML, Eckardt JR, Burris HA, et al: Irinotecan (CPT-11) as second-line therapy for patients with 5-FU-refractory colorectal cancer (abstract). Proc Am Soc Clin Oncol 13:A578, 1994.

40. Pitot HC, Wender D, O'Connell MJ, et al: A phase II trial of CPT-11 (irinotecan) in patients with metastatic colorectal carcinoma: A NCCTG study (abstract). Proc Am Soc Clin Oncol 13:A573, 1994.

41. Bugat R, Suc E, Rougier P, et al: CPT-11 (irinotecan) as second-line therapy in advanced colorectal cancer (CRC): Preliminary results of multicentric phase II study (abstract). Proc Am Soc Clin Oncol 13:A586, 1994.

42. Rougier P, Culine S, Bugat R, et al: Multicentric phase II study of first line CPT-11 (irinotecan) in advanced colorectal cancer (CRC): Preliminary results (abstract). Proc Am Soc Clin Oncol 13:A585, 1994.

43. Tsuda H, Takatsuki K, Ohno R, et al: Treatment of adult T-cell leukaemia-lymphoma with irinotecan hydrochloride (CPT-11): CPT-11 Study Group on Hematological Malignancy. Br J Cancer 70(4):771-774, 1994.

44. Shinkai T, Arioka H, Kunikane H, et al: Phase I clinical trial of irinotecan (CPT-11), 7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxy-camptothecin, and cisplatin in combination with fixed dose of vindesine in advanced non-small-cell lung cancer. Cancer Res 54(10):2636-2642, 1994.

45. Mori K, Suga U, Kishiro I, et al: A phase I study of CPT-11 and cisplatin (5-day continuous infusion) for advanced non-small-cell lung cancer (abstract). Proc Am Soc Clin Oncol 13:A1234, 1994.

46. Kambe M, Wakui A, Nakao I, et al: A late phase II study of irinotecan (CPT-11) in patients (pts) with advanced gastric cancer (abstract). Proc Am Soc Clin Oncol 12:A584, 1993.

47. Rowinsky E, Sartorius S, Grochow L, et al: Phase I and pharmacologic study of topotecan, an inhibitor of topoisomerase I, with granulocyte colony-stimulating factor (G-CSF): Toxicologic differences between concurrent and post-treatment G-CSF administration (abstract). Proc Am Soc Clin Oncol 11:A284, 1992.

48. Slichenmyer W, Chen TL, Donehower R, et al: Clinical pharmacology of topotecan in cancer patients with renal or hepatic dysfunction (abstract). Proc Am Soc Clin Oncol 13:A363, 1994.

49. Schiller JH, Kim K, Johnson D: Phase II study of topotecan in extensive stage small-cell lung cancer (abstract). Proc Am Soc Clin Oncol 13:A1093, 1994.

50. Robert F, Wheeler RH, Molthrop DC, et al: Phase II study of topotecan in advanced head and neck cancer: Identification of an active new agent (abstract). Proc Am Soc Clin Oncol 13:A905, 1994.

51. Sugarman SM, Ajani JA, Daugherty K, et al: A phase Ill trial of topotecan (TPT) for the treatment of advanced, measurable colorectal ca (abstr). Proc Am Soc Clin Oncol 13:A686, 1994.

52. Sugarman SM, Pazdur R, Daugherty K, et al: A phase II trial of topotecan (TPT) for the treatment of unresectable pancreatic cancer (abstract). Proc Am Soc Clin Oncol 13:A684, 1994.

53. Eisenhauer EA, Wainman N, Boos G, et al: Phase II trials of topotecan in patients (pts) with malignant glioma and soft tissue sarcoma (abstract). Proc Am Soc Clin Oncol 13:A488, 1994.

54. Perez-Soler R, Glisson BS, Kane J, et al: Phase II study of topotecan in patients with non-small-cell lung cancer (NSCLC) previously untreated (abstract). Proc Am Soc Clin Oncol 13:A1223, 1994.

55. Ardizzoni A, Hansen H, Dombernowsky P, et al: Phase II study of topotecan in pretreated small-cell lung cancer (SCLC) (abstract). Proc Am Soc Clin Oncol 13:A1116, 1994.

56. Lynch TJ Jr, Kalish L, Strauss G, et al: Phase II study of topotecan in metastatic NSCLC. J Clin Oncol 12(2):347-352, 1994.

57. Ilson D, Motzer RJ, O'Moore P, et al: A phase II trial of topotecan in advanced renal cell carcinoma (abstract). Proc Am Soc Clin Oncol 12:A779, 1993.

58. Kudelka A, Edwards C, Freedman R, et al: An open phase II study to evaluate the efficacy and toxicity of topotecan administered iv as 5 daily infusions every 21 days to women with advanced epithelial ovarian carcinoma (abstract). Proc Am Soc Clin Oncol 12:A821, 1993.

59. Rowinsky E, Grochow L, Kaufmann S, et al: Sequence-dependent effects of topotecan (T) and cisplatin (C) in a phase I and pharmacokinetic (PK) study (abstract). Proc Am Soc Clin Oncol 13:A361, 1994.

60. Graham MV, Jahanzeb M, Dresler C, et al: Preliminary results of a phase I study of topotecan plus thoracic radiotherapy for locally advanced non-small-cell lung cancer (NSCLCA) (abstract). Proc Am Soc Clin Oncol 13:A1132, 1994.

61. Tolcher AW, O'Shaughnessy JA, Weiss RB, et al: A phase I study of topotecan (a topoisomerase I inhibitor) in combination with doxorubicin (a topo-isomerase II inhibitor) (abstract). Proc Am Soc Clin Oncol 13:A422, 1994.

62. Eckardt JR, Burris HA, Von Hoff DD, et al: Measurement of tumor topoisomerase I and II levels during the sequential administration of topotecan and etoposide (abstract). Proc Am Soc Clin Oncol 13:A358, 1994.

63. Lilenbaum RC, Rosner GL, Ratain MJ, et al: Phase I study of Taxol and topotecan in patients with advanced solid tumors (CALGB 9362) (abstract). Proc Am Soc Clin Oncol 13:A319, 1994.

64. Eckardt JR, Von Hoff DD, Rinaldi DA, et al: Phase I trial of cisplatin followed by topotecan in patients with untreated non-small-cell lung cancer (abstract). Fifth Conference on DNA Topoisomerases in Therapy 48:P47, 1994.

65. Miller AA, Hargis JB, Lilenbaum RC, et al: Phase I study of topotecan and cisplatin in patients with advanced solid tumors: A cancer and leukemia group B study. J Clin Oncol 12(12):2743-2750, 1994.

66. Creemers GJ, Schellens JH, Beijnen JH, et al: Bioavailability of oral topotecan: A new topoisomerase I inhibitor (abstract). Proc Am Soc Clin Oncol 13:A324, 1994.

67. Cassidy J, Kaye SB: New drugs in clinical development in Europe. Hematol Oncol Clin North Am 8(2):289-303, 1994.

68. Wanders J, Schrijvers D, Bruntsch U, et al: The EORTC-ECTG experience with acute hypersensitivity reactions (HSR) in taxotere studies. Proc Am Soc Clin Oncol 12:73, 1993.

69. Wanders J, Van Oosterom A, Gore M, et al: Taxotere toxicity-protective effects of premedication. Eur J Cancer 29A(6):S206, 1993.

70. Eisenhauer EA, Lu F, Muldal A, et al: Predictors and treatment of docetaxel toxic effects. Ann Oncol 5(5):202, 1994.

71. Oulid-Aissa D, Behar A, Speilmann M, et al: Management of fliud retention syndrome in patients with taxotere (docetaxel): Effect of premedication. Proc Am Soc Clin Oncol 13:465, 1994.

72. New P: Neurotoxicity of taxotere. Proc Am Assoc Cancer Res 34:233, 1993.

73. Bruno R, Cosson V, Vergniol JC, et al: Taxotere population pharmacokinetics. Proc Am Assoc Cancer Res 34:234, 1993.

74. De Valeriola D, Brassinne C, Gaillard C, et al: Study of excretion balance, metabolism, and protein binding of 14C radiolabelled taxotere (RP 56976, NSC 628503) in cancer patients. Proc Am Assoc Cancer Res 34:373, 1993.

75. Ten Bokkel Huinink WW, Prove AM, Piccart M, et al: A phase II trial with docetaxel (Taxotere) in second line treatment with chemotherapy for advanced breast cancer. A study of the EORTC Early Clinical Trials Group. Ann Oncol 5(6):527-532, 1994.

76. Valero V, Walters R, Theriault R, et al: Phase II study of docetaxel (Taxotere) in anthracycline-refractory metastatic breast cancer (ARMBC) (abstract). Proc Am Soc Clin Oncol 13:A1636, 1994.

77. Dieras V, Fumoleau P, Chevallier B, et al: Second EORTC-clinical screening group (CSG) phase II trial of Taxotere (docetaxel) as first line chemotherapy (CT) in advanced breast cancer (ABC) (abstract). Proc Am Soc Clin Oncol 13:A115, 1994.

78. Trudeau ME, Eisenhauer E, Lofters W, et al: Phase II study of Taxotere as first-line chemotherapy for metastatic breast cancer: A National Cancer Institute of Canada clinical trials group (NCIC CTG) study (abstract). Proc Am Soc Clin Oncol 12:A59, 1993.

79. Seidman AD, Hudis C, Crown JP, et al: Phase II evaluation of Taxotere (RP56976, NSC628503) as initial chemotherapy for metastatic breast cancer (abstract). Proc Am Soc Clin Oncol 12:A52, 1993.

80. Cerny T, Kaplan S, Pavlidis N et al: Docetaxel (Taxotere) is active in non-small-cell lung cancer: A phase II trial of the EORTC Early Clinical Trials Group (ECTG). Br J Cancer 70(2):384-387, 1994.

81. Rigas JR, Francis PA, Kris MG, et al: Phase II trial of Taxotere in non-small-cell lung cancer (NSCLC) (abstract). Proc Am Soc Clin Oncol 12:A1121, 1993.

82. Burris H, Eckardt J, Fields S, et al: Phase II trials of Taxotere in patients with non-small-cell lung cancer (abstract). Proc Am Soc Clin Oncol 12:A1116, 1993.

83. Fossella FV, Lee JS, Shin DM, et al: Taxotere (docetaxel: DTXL), an active agent for platinum-refractory non-small-cell lung cancer (NSCLC): Preliminary report of a phase II study (abstr). Proc Am Soc Clin Oncol 3:A1115, 1994.

84. Watanabe K, Yokoyama A, Furuse K et al: Phase 11 trial of docetaxel in previously untreated non-small cell lung cancer (NSCLC) (abstract). Proc Am Soc Clin Oncol 13:A1095, 1994.

85. Fossella FV, Lee JS, Murphy WK, et al: Phase II study of docetaxel for recurrent or metastatic non-small-cell lung cancer. J Clin Oncol 12(6):1238-1244, 1994.

86. Francis P, Hakes T, Schneider J, et al: Phase II study of docetaxel (Taxotere) in advanced platinum-refractory ovarian cancer (ca) (abstract). Proc Am Soc Clin Oncol 13:A825, 1994.

87. Kavanagh J, Kudelka A, Freedman R, et al: Taxotere (docetaxel): Activity in platin refractory ovarian cancer and amelioration of toxicity (abstr). Proc Am Soc Clin Oncol 13:A732, 1994.

88. Aapro M, Pujade-Lauraine E, Lhomme C, et al: Phase II study of Taxotere (T) in ovarian cancer: EORTC Clinical Screening Group (CSG) (abstract). Proc Am Soc Clin Oncol 12:A809, 1993.

89. Sadan S, Bajorin D, Amsterdam A, et al: Docetaxel in patients with advanced transitional cell cancer (TCC) who failed cisplatin-based chemotherapy: A phase II trial (abstract). Proc Am Soc Clin Oncol 13:A761, 1994.

90. Einzig AI, Schuchter LM, Wadler S, et al: Phase II trial of Taxotere (RP 56976) in patients (pts) with metastatic melanoma previously untreated with cytotoxic chemotherapy (abstract). Proc Am Soc Clin Oncol 13:A1345, 1994.

91. Bedikian A, Legha S, Eton O, et al: Phase II trial of docetaxel (Taxotere, RP 56976) in patients with advanced cutaneous malignant melanoma (ACMM) previously untreated with chemo-Rx. Proc Am Assoc Cancer Res 35:A512, 1994.

92. Rougier P, De Forni M, Adenis A, et al: Phase II study of Taxotere (RP56976, docetaxel) in pancreatic adenocarcinoma (PAC) (abstract). Proc Am Soc Clin Oncol 13:A587, 1994.

93. Mertens WC, Eisenhauer EA, Jolivet J, et al: Docetaxel in advanced renal carcinoma. A phase II trial of the National Cancer Institute of Canada Clinical Trials Group. Ann Oncol 5(2):185-187, 1994.

94. Van Hoesel QG, Verweij J, Catimel G, et al: Phase II study with docetaxel (Taxotere) in advanced soft tissue sarcomas. Ann Oncol 5(6):539-542, 1994.

95. Dreyfuss A, Clark J, Norris C, et al: Taxotere (TXTR) for advanced, incurable squamous cell carcinoma of the head and neck (abstract). Proc Am Soc Clin Oncol 13:A931, 1994.

96. Catimel G, Verweij J, Mattijssen V, et al: Docetaxel (Taxotere): An active drug for the treatment of patients with advanced squamous cell carcinoma of the head and neck: EORTC Early Clinical Trials Group. Ann Oncol 5(6):533-537, 1994.

97. Clark T, Kemeny N, Conti JA, et al: Phase II trial of docetaxel (Taxotere, RP56976) in previously untreated patients (pts) with advanced colorectal cancer (CRC) (abstract). Proc Am Soc Clin Oncol 13:A635, 1994.

98. Pazdur R, Lassere Y, Soh L, et al: Phase II trial of docetaxel (Taxotere) in metastatic colorectal carcinoma. Ann Oncol 5(6):468-470, 1994.

99. Sternberg CN, Ten Bokkel Huinink WW, Smyth JF, et al: Docetaxel (Taxotere), a novel taxoid, in the treatment of advanced colorectal carcinoma: An EORTC Early Clinical Trials Group Study. Br J Cancer 70(2):376-379, 1994.

100. Sulkes A, Smyth J, Sessa C, et al: Docetaxel (Taxotere) in advanced gastric cancer: Results of a phase II clinical trial: EORTC Early Clinical Trials Group. Br J Cancer 70(2):380-383, 1994.

101. Eckardt JR, Von Hoff DD: New drugs in clinical development in the United States. Hematol Oncol Clin North Am 8(2):305-332, 1994.

102. Boven E, Erkelens CA, Pinedo HM, et al: The new cytidine analog gemcitabine (GEM) has schedule-rather than dose-related activity in human tumor xenografts. Proc Am Assoc Cancer Res 32:A2270, 1991.

103. Lund B, Hansen OP, Theilade K, et al: Phase II study of gemcitabine (2¢2¢-difluorodeoxycytidine) in previously treated ovarian ca patients. J Natl Cancer Inst 86(20):1530-1533, 1994.

104. Catimel G, Vermorken JB, Clavel M, et al: A phase II study of gemicitabine (LY188011) in patients with advanced squamous cell carcinoma of the head and neck: EORTC Early Clinical Trials Group. Ann Oncol 5(6):543-547, 1994.

105. Anderson H, Lund B, Bach F, et al: Single-agent activity of weekly gemcitabine in advanced non-small-cell lung cancer: A phase II study. J Clin Oncol 12(9):1821-1826, 1994.

106. Sessa C, Aamdal S, Wolff I, et al: Gemcitabine in patients with advanced malignant melanoma or gastric cancer: Phase II studies of the EORTC Early Clinical Trials Group. Ann Oncol 5(5):471-472, 1994.

107. Abratt RP, Bexwoda WR, Falkson G, et al: Efficacy and saftey profile of gemcitabine in non-small-cell lung cancer: A phase II study. J Clin Oncol 12(8):1535-1540, 1994.

108. Cormier Y, Eisenhauer E, Muldal A, et al: Gemcitabine is an active new agent in previously untreated extensive small cell lung cancer (SCLC): A study of the National Cancer Institute of Canada Clinical Trials Group. Ann Oncol 5(3):283-285, 1994.

109. Negoro S, Fukuoka M, Kurita Y, et al: Results of phase II studies of gemcitabine in non-small-cell lung cancer (NSCLC). Proc Am Soc Clin Oncol 13:A1239, 1994.

110. Fossella FV, Lippman S, Pang A, et al: Phase I/II study of gemcitabine (G) by 30 minute weekly iv infusion x 3 weeks every 4 weeks for non-small-cell lung cancer (NSCLC). Proc Am Soc Clin Oncol 12:A1082, 1993.

111. Christman K, Kelsen D, Saltz L, et al: Phase II trial of gemcitabine in patients with advanced gastric cancer. Cancer 73(1):5-7, 1994.

112. Carmichael J, Possinger K, Philip P, et al: Difluorodeoxycytidine (gemcitabine): A phase II study in patients with advanced breast cancer. Proc Am Soc Clin Oncol 12:A57, 1993.

113. Mertens WC, Eisenhauer EA, Moore M, et al: Gemcitabine in advanced renal cell carcinoma: A phase II study of the National Cancer Institute of Canada Clinical Trial Group. Ann Oncol 4(4):331-332, 1993.

114. Shepherd FA, Gatzemeier U, Gotfried M, et al: An extended phase II study of gemcitabine in non-small-cell lung cancer (NSCLC). Proc Am Soc Clin Oncol 12:A1096, 1993.

115. Carmichael J, Jink U, Rusell RC, et al: Phase II study of gemcitabine in patients with advanced pancreated cancer. Proc Am Soc Clin Oncol 12:A698, 1993.

116. Moore DF Jr, Pazdur R, Daugherty K, et al: Phase II study of gemcitabine in advanced colorectal adenocarcinoma. Invest New Drugs 10(4):323-325, 1992.

117. Lund B, Ryberg M, Meidal P, et al: A phase II study of gemcitabine in non-small-cell lung cancer (NSCLC) using a twice-weekly schedule. Ann Oncol 3(suppl 5):31, 1992.

118. Weissbach L, de Mulder P, Osieka R, et al: Phase II study of gemcitabine in renal cancer. Proc Am Soc Clin Oncol 11:A689, 1992.

119. Fink U, Molle B, Daschner H, et al: Phase II study of gemcitabine in metastatic colorectal cancer. Proc Am Soc Clin Oncol 11:A507, 1992.

120. Casper ES, Green MR, Brown TD, et al: Phase II trial of gemcitabine (2¢2¢-difluorodeoxycytidine) in patients with pancreatic cancer. Proc Am Soc Clin Oncol 10:A440, 1991.

121. Peters G, Tanis B, Clavel M, et al: Pharmacokinetics of gemcitabine (LY188011) (difluoro-deoxycytidine) administered every two weeks in a phase I study. Proc Am Assoc Cancer Res 31:180, 1990.

122. Allerheiligen S, Johnson R, Hatcher B, et al: Gemcitabine pharmacokinetics are influenced by gender, body surface area (BSA), and duration of infusion. Proc Am Soc Clin Oncol 13:A338, 1994.

123. Andersen JS, Burris HA, Casper E, et al: Development of a new system for assessing clinical benefit for patients with advanced pancreatic cancer. Proc Am Soc Clin Oncol 13:A1600, 1994.

124. Heidelberger C, Chandhari NK, Dannenberg P, et al: Fluorinated pyrimidines: A new class of tumor inhibitory compounds. Nature 179:663-666, 1957.

125. Buroker T, Padilla A, Groppe C, et al: Phase II evaluation of Ftorafur in previously untreated colorectal cancer, a Southwest Oncology Group study. Cancer 44:48-51, 1979.

126. Ansfield FJ, Kallas GJ, Singson JP: Phase I-II studies of oral Tegafur (Ftorafur). J Clin Oncol 1(2):107-110, 1983.

127. Okeda R, Shibutani M, Matsuo T, et al: Experimental neurotoxicity of 5-fluorouracil and its derivatives is due to poisoning by the monofluorinated organic metabolites, monofluoroacetic acid and -fluoro- alanine. Acta Neuropathol 81:66-73, 1990.

128. Malik ST, Talbot D, Clarke PI, et al: Phase II trial of UFT in advanced colorectal and gastric cancer. Br J Cancer 62(6):1023-1025, 1990.

129. Gonzalez Baron M, Feliu J, Ordonez A, et al: Phase I study of UFT plus leucovorin in advanced colorectal cancer: A double modulation proposal. Anticancer Res 13(3):759-762, 1993.

130. Pazdur R, Lassere Y, Rhodes V, et al: Phase II trial of Uracil and Tegafur plus oral leucovorin: An effective oral regimen in the treatment of metastatic colorectal carcinoma. J Clin Oncol 2(11):2296-2300, 1994.

131. Cao S, Rustum YM, Spector T: 5-Ethynyluracil (776C85): Modulation of 5-fluorouracil efficacy and therapeutic index in rats bearing advanced colorectal carcinoma. Cancer Res 54:1507-1510, 1994.

132. Davis ST, Joyner SS, Baccanari DP, et al: 5-Ethynyluracil (5-EU, 776C85) improves the pharmacokinetics of 5-fluorouracil (5-FU) in rats dosed with ftorafur (FT). Proc Amer Assoc Cancer Res 35:A1912, 1994.

133. Davis ST, Joyner SS, Baccanari DP, et al: 5-Ethynyluracil (776C85): Protection from 5-fluorouracil-induced neurotoxicity in dogs. Biochem Pharmacol 48(2):233-236, 1994.

134. Peters GJ, van der Wilt CL, van Groeningen K, et al: Thymidylate synthase inhibition after administration of fluorouracil with or without leucovorin in colon cancer patients: Implications for treatment with fluorouracil. J Clin Oncol 12(10):2035-2042, 1994.

135. Jackman AL, Taylor GA, Gibson W, et al: ICI D1694, a quinazoline antifolate thymidylate synthase inhibitor that is a potent inhibitor of L1210 tumor cell growth in vitro and in vivo: A new agent for clinical study. Cancer Res 51:5579-5586, 1991.

136. Weiss GR, Eckardt JR, Eckhardt SG, et al: New anticancer agents, in Pinteto HM, Longo DL, Chabner BA (eds): Cancer Chemotherapy and Biological Response Modifiers Annual 16. Amsterdam, Elsevier Science, 1995 (in press).

137. Rinaldi DA, Burris HA, Dorr FA, et al: A phase I evaluation of the novel thymidylate synthase inhibitor LY231514, in patients with advanced solid tumors. Proc Amer Soc Clin Oncol 13:A430, 1994.

138. Yanase T, Tamura M, Fujita K, et al: Inhibitory effect of angiogenesis inhibitor TNP-470 on tumor growth and metastasis of human cell lines in vitro and in vivo. Cancer Res 53:2566-2570, 1993.

139. Brem H, Grasser I, Grosfeld J, et al: The combination of antiangiogenic agents to inhibit primary tumor growth and metastasis. J Ped Surg 28:1253-1257, 1993.

140. Nakamura S, Sakurada S, Salahuddin S, et al: Inhibition of development of Kaposi's sarcoma-related lesions by a bacterial cell wall complex. Science 255:1437-1440, 1989.

141. Eckhardt SG, Burris HA, Eckardt JR, et al: Phase I assessment of the novel angiogenesis inhibitor DS4152 (tecogalan sodium). Proc Am Soc Clin Oncol 13:55, 1994.

142. Staddon A, Henry D, Bonnem E: A randomized dose finding study of recombinant platelet factor 4 (rPF4) in cutaneous AIDS-related Kaposi's sarcoma (KS). Proc Am Soc Clin Oncol 13:50, 1994.

143. Belman N, Lipton A, Harvey H, et al: rhuPF4-phase I study of an angiogenesis inhibitor in metastatic colon cancer (MCC). Proc Am Soc Clin Oncol 13:221, 1994.

144. Yamamoto T, Sudo K, Fujita T, et al: Significant inhibition of endothelial cell growth in tumor vasculature by an angiogenesis inhibitor, TNP-470 (AGM-1470). Anticancer Res 14:1-4, 1994.

145. Yanase T, Tamura M, Fujita K, et al: Inhibitory effect of angiogenesis inhibitor TNP-470 on tumor growth and metastasis of human cell lines in vitro and in vivo. Cancer Res 53:2566-2570, 1993.

146. Pluda J, Wyvill W, Figg S, et al: A phase I study of an angiogenesis inhibitor, TNP-470 (AGM-1470), administered to patients (PTS) with HIV-associated Kaposi's sarcoma (KS). Proc Am Soc Clin Oncol 13:51, 1994.

147. Zukiwski A, Gutterman C, Bui C, et al: Phase I trial of the angiogenesis inhibitor TNP-470 (AGM-1470) in patients with androgen independent prostate cancer (AI PCa). Proc Am Soc Clin Oncol 13:252, 1994.

Related Videos
Interim data reveal favorable responses in patients with low-grade serous ovarian cancer treated with avutometinib plus defactinib, according to Susana N. Banerjee, MD.
Treatment with mirvetuximab soravtansine appears to produce a 3-fold improvement in objective response rate vs chemotherapy among patients with folate receptor-α–expressing, platinum-resistant ovarian cancer in the phase 3 MIRASOL trial.
PRGN-3005 autologous UltraCAR-T cells appear well-tolerated and decreases tumor burden in a population of patients with advanced platinum-resistant ovarian cancer.
An expert from Dana-Farber Cancer Institute discusses findings from the final overall survival analysis of the phase 3 ENGOT-OV16/NOVA trial.