there are no bad anticancer agents, only bad clinical trial ......vol. 4, 1079 -1086, may 1998...

9
Vol. 4, 1079 -1086, May 1998 Clinical Cancer Research 1079 Special Lecture There Are No Bad Anticancer Agents, Only Bad Clinical Trial Designs-Twenty-first Richard and Hinda Rosenthal Foundation Award Lecture1 Daniel D. Von Hoff 2,3 Institute for Drug Development, Cancer Therapy and Research Center, and Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78245 Abstract Unfortunately, the vast majority (90%) of new antican- cer agents designed in the laboratory never make it into routine clinical use. The hypothesis of this lecture is that many new agents fail in the clinic because the appropriate clinical trial(s) that could exploit the attributes of the new agent are not performed. An appreciation that both bench and clinical investigations are difficult endeavors should aid in improving clinical trial designs and give the best chance for new agents to be added to our therapeutic armamen- tarium. Introduction A very common story is that a basic scientist works for years and years to develop a new agent with a unique mecha- nism of action to treat tumor cells; but that new agent never makes it into clinical trials, or if it does, it almost always fails to make an impact on the disease. The hypothesis of today’s lecture is that many new agents fail because the appropriate clinical trials to exploit the at- tributes of the new agent have not been performed. This is because of a basic uncoupling or disconnection between the bench scientist, who has worked long and hard on his or her hypothesis but who may not understand the difficulties of and creativity required for clinical trials, and the clinical scientist, who may not understand how difficult the basic discovery has been and how careful one must be before abandoning the new concept or new agent from the bench. The magnitude of the disconnection between the bench scientist and the bedside clinician cannot be assessed solely by how many new agents don’t make it into general use in the oncology community. The percentage of agents, however, that do go to clinical trial and are adopted for clinical use is disap- Received 2/3/98; accepted 3/4/98. 1 Presented at the 88th Annual Meeting of the American Association for Cancer Research, April 15, 1997, San Diego. CA. 2 To whom requests for reprints should be addressed, at Institute for Drug Development, Cancer Therapy and Research Center, 14960 Omicron Drive, San Antonio, TX 78245. Phone: (210) 677-3880; Fax: (2 10) 677-0058; E-mail: [email protected]. 3 Dr. Von Hoff is a founder and minority owner of ILEX Oncology, which is working on clinical trials with MGBG and DFMO. He is also a member of the Scientific Advisory Board for Eli Lilly, which owns and markets gemcitabine. pointingly low. Fig. 1 details the number of new agents brought into clinical trials from 1975 to 1994 and the number of those agents eventually approved for clinical use. Overall, there were 280 new agents brought into Phase I (dose-finding) clinical trials in patients. Only 29 of them (10%) were eventually ap- proved for use in patients. There are three main reasons that emerge in examining why the other 90% of new agents don’t make it into routine use. These include: (a) the toxicities of the agent were too great; (b) there was a lack of efficacy; and (c) no attention was paid to the mechanism of action of the compound when the clinical trials were designed and conducted. In the discussion below, we will explore all three of these major reasons why new agents don’t make it into general clinical use. In addition, examples will be given of how to bypass these problems so that a greater percentage of agents brought into clinical trials will actually make it to general use. Why Compounds Don’t Make It-Toxicities Table 1 gives examples of severe toxicities that halted the development of several new anticancer agents. These toxicities basically stopped the development of the new agents in their tracks, in most instances, with a complete suspension of their clinical trials. However, because of the clinical activity of the agent that had been seen early on, e.g. , responses to the agent paclitaxel in patients with a variety of malignancies, clinicians persisted in trying to find a way to circumvent the toxicity of paclitaxel. The thesis is that one can circumvent toxicities of the new agent if the drug has clinical antitumor activity. Table 1 also details the methods that were used to circumvent toxicities for each of the agents. The observation of our drug development team in San Antonio, TX, is that in dealing with the phenomenon of dose- limiting toxicities, stopping the drug and then the clinician finding a way to circumvent those toxicities, we have devised the life cycle of a new anticancer drug as shown in Fig. 2 (1). As can be seen in the figure, a drug is discovered and brought into clinical testing. Almost inevitably, a crisis in the development of the agent occurs in which either a severe toxicity is noted with the agent or no antitumor activity is noted with the agent. The agent then undergoes programmed drug death, which we like to call “pharmacoptosis.” Fortunately, methods can sometimes be developed to circumvent the toxicity of the new agent, and the agent then emerges from the state of pharmacoptosis and is rediscovered or reborn. As is noted in Table 1, this ability to reverse pharmacoptosis is a very common phenomenon. An Example of Recovery from Pharmacoptosis Because of Toxicity-Mitoguazone. A recent example of recovery from pharmacoptosis is the redevelopment of mitoguazone. Mitoguazone was synthesized in 1898 by Thiele et a!. (2). It also has the names MGBG, methylglyoxal bis(guanylhydrazone), Research. on June 13, 2021. © 1998 American Association for Cancer clincancerres.aacrjournals.org Downloaded from

Upload: others

Post on 30-Jan-2021

5 views

Category:

Documents


0 download

TRANSCRIPT

  • Vol. 4, 1079 -1086, May 1998 Clinical Cancer Research 1079

    Special Lecture

    There Are No Bad Anticancer Agents, Only Bad Clinical Trial

    Designs-Twenty-first Richard and Hinda Rosenthal

    Foundation Award Lecture1

    Daniel D. Von Hoff 2,3Institute for Drug Development, Cancer Therapy and ResearchCenter, and Department of Medicine, University of Texas HealthScience Center at San Antonio, San Antonio, Texas 78245

    Abstract

    Unfortunately, the vast majority (90%) of new antican-

    cer agents designed in the laboratory never make it into

    routine clinical use. The hypothesis of this lecture is that

    many new agents fail in the clinic because the appropriate

    clinical trial(s) that could exploit the attributes of the new

    agent are not performed. An appreciation that both bench

    and clinical investigations are difficult endeavors should aid

    in improving clinical trial designs and give the best chance

    for new agents to be added to our therapeutic armamen-

    tarium.

    IntroductionA very common story is that a basic scientist works for

    years and years to develop a new agent with a unique mecha-

    nism of action to treat tumor cells; but that new agent never

    makes it into clinical trials, or if it does, it almost always fails to

    make an impact on the disease.

    The hypothesis of today’s lecture is that many new agents

    fail because the appropriate clinical trials to exploit the at-

    tributes of the new agent have not been performed. This is

    because of a basic uncoupling or disconnection between the

    bench scientist, who has worked long and hard on his or her

    hypothesis but who may not understand the difficulties of and

    creativity required for clinical trials, and the clinical scientist,

    who may not understand how difficult the basic discovery has

    been and how careful one must be before abandoning the new

    concept or new agent from the bench.

    The magnitude of the disconnection between the bench

    scientist and the bedside clinician cannot be assessed solely by

    how many new agents don’t make it into general use in the

    oncology community. The percentage of agents, however, that

    do go to clinical trial and are adopted for clinical use is disap-

    Received 2/3/98; accepted 3/4/98.1 Presented at the 88th Annual Meeting of the American Association for

    Cancer Research, April 15, 1997, San Diego. CA.

    2 To whom requests for reprints should be addressed, at Institute forDrug Development, Cancer Therapy and Research Center, 14960Omicron Drive, San Antonio, TX 78245. Phone: (210) 677-3880; Fax:(2 10) 677-0058; E-mail: [email protected] Dr. Von Hoff is a founder and minority owner of ILEX Oncology,

    which is working on clinical trials with MGBG and DFMO. He is alsoa member of the Scientific Advisory Board for Eli Lilly, which owns

    and markets gemcitabine.

    pointingly low. Fig. 1 details the number of new agents brought

    into clinical trials from 1975 to 1994 and the number of those

    agents eventually approved for clinical use. Overall, there were

    280 new agents brought into Phase I (dose-finding) clinical

    trials in patients. Only 29 of them (10%) were eventually ap-

    proved for use in patients.

    There are three main reasons that emerge in examining

    why the other 90% of new agents don’t make it into routine use.

    These include: (a) the toxicities of the agent were too great; (b)

    there was a lack of efficacy; and (c) no attention was paid to the

    mechanism of action of the compound when the clinical trials

    were designed and conducted.

    In the discussion below, we will explore all three of these

    major reasons why new agents don’t make it into general

    clinical use. In addition, examples will be given of how to

    bypass these problems so that a greater percentage of agents

    brought into clinical trials will actually make it to general use.

    Why Compounds Don’t Make It-ToxicitiesTable 1 gives examples of severe toxicities that halted the

    development of several new anticancer agents. These toxicities

    basically stopped the development of the new agents in their

    tracks, in most instances, with a complete suspension of their

    clinical trials. However, because of the clinical activity of the

    agent that had been seen early on, e.g. , responses to the agent

    paclitaxel in patients with a variety of malignancies, clinicians

    persisted in trying to find a way to circumvent the toxicity of

    paclitaxel. The thesis is that one can circumvent toxicities of the

    new agent if the drug has clinical antitumor activity. Table 1

    also details the methods that were used to circumvent toxicities

    for each of the agents.

    The observation of our drug development team in San

    Antonio, TX, is that in dealing with the phenomenon of dose-

    limiting toxicities, stopping the drug and then the clinician

    finding a way to circumvent those toxicities, we have devised

    the life cycle of a new anticancer drug as shown in Fig. 2 (1). As

    can be seen in the figure, a drug is discovered and brought into

    clinical testing. Almost inevitably, a crisis in the development of

    the agent occurs in which either a severe toxicity is noted with

    the agent or no antitumor activity is noted with the agent. The

    agent then undergoes programmed drug death, which we like to

    call “pharmacoptosis.” Fortunately, methods can sometimes be

    developed to circumvent the toxicity of the new agent, and the

    agent then emerges from the state of pharmacoptosis and is

    rediscovered or reborn. As is noted in Table 1, this ability to

    reverse pharmacoptosis is a very common phenomenon.

    An Example of Recovery from Pharmacoptosis Because

    of Toxicity-Mitoguazone. A recent example of recovery

    from pharmacoptosis is the redevelopment of mitoguazone.

    Mitoguazone was synthesized in 1898 by Thiele et a!. (2). It also

    has the names MGBG, methylglyoxal bis(guanylhydrazone),

    Research. on June 13, 2021. © 1998 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • 40

    .nC.)

    � 30rI)

    .�

    I-

    5)

    75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94

    Year

    1080 There Are No Bad Anticancer Drugs

    Fig. 1 Total number of differ-ent agents in Phase I trials per

    year (#{149}),and number of thoseagents eventually approved (El).

    methyl-GAG, and NSC-32946. The structure of mitoguazone is

    shown in Fig. 3. One of the intriguing aspects of mitoguazone is

    that it has a unique mechanism of action as an inhibitor of

    S-adenosylmethionine decarboxylase (3, 4). Some investigators

    have believed that it is also a mitochondrial interactive agent (5).

    It should be remembered that compounds with new and uniquemechanisms of action have a better chance for non-cross-resis-

    tance with the antineoplastics already in clinical use.

    Mitoguazone was first given to patients in the early l960s,

    usually on a chronic (e.g., daily X 30 days) schedule (6, 7).

    Antitumor activity was noted in patients with leukemia and

    lymphoma (6- 8). However, the major toxicity of mucositis (and

    other severe toxicities) caused all clinical trials with the agent tobe stopped. Therefore, despite clinical activity, all clinical trials

    with mitoguazone were discontinued (the drug underwent phar-

    macoptosis).

    The key to the rebirth of mitoguazone was new scientificknowledge from pharmacokinetic studies performed in the late

    1970s and early l980s (and reconfirmed recently) that mitogua-

    zone had an extremely long plasma half-life of >5 days (9-1 1).With this new understanding, it was clear that the schedule of

    daily administration of mitoguazone was causing an accumula-

    tion of mitoguazone that could have been responsible for the

    side-effect profile (severe mucositis and myelosuppression sec-

    ondary to toxicity on rapidly turning over cells in the mucous

    membranes). Based on this new knowledge, weekly and every

    other week schedules of administration were devised (12, 13).

    Using these dosing schedules, mucositis became an unusual side

    effect with the agent, and antitumor activity was noted in pa-

    tients with Hodgkin’s disease, non-Hodgkin’s lymphoma, head

    and neck cancer, endometrial cancer, prostate cancer, and

    Table 1 Agents in which severe toxicities halted their developmentand methods used to circumvent those toxicities (and reverse

    pharmacoptosis)

    Agent Toxicity

    Method tocircumvent

    toxicity

    Doxorubicin/ Congestive heart Dose limitation,Daunomycin failure dexrazoxane

    Vincristine Neurotoxicity Dose limitationCisplatin Renal toxicity,

    ototoxicity emesisHydration,

    antiemeticsPaclitaxel Anaphylactic shock Steroids and other

    premedicationDocetaxel Fluid retention SteroidsCPT-l 1 Diarrhea AntidiarrhealsFludarabine Blindness Dose limitation

    phosphate

    esophageal cancer (12-16). Despite that activity, there was still

    little interest in mitoguazone until the emergence of non-Hodgkin’s lymphoma in patients with AIDS.

    AIDS-related non-Hodgkin’s lymphoma was first reportedin 1982. It is usually a disease with aggressive histologies

    (large-cell immunoblastic and small-cell non-cleaved), has a

    high incidence of central nervous system involvement, accounts

    for 12-16% of all AIDS-related deaths, and there is no standard

    treatment for patients who have progressed on first-line therapy

    (17, 18). The average survival for patients who have progressed

    on first-line therapy is 63 days (19). Polyamines are elevated in

    patients with lymphoma 4.0-5.3-fold compared with normal

    volunteers (20, 21); therefore, an agent like mitoguazone, which

    Research. on June 13, 2021. © 1998 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • Discovery(Birth)

    Rediscovery(Rebirth)

    Fig. 3 Structure of mitoguazone.

    Fig. 2 The life cycle of a new anticancer drug (reprinted with permis-

    sion from Annals of Oncology).

    100w

    �g 750-�:

    250-

    25 30 35 40 45

    Fig. 4 Antitumor activity of the hypothetical new agent difungomuc-

    tane ( #{149}) in an in vivo tumor growth system compared with controlgrowth (0) of the tumor.

    Clinical Cancer Research 1081

    Clinicaltesting

    �ll�p�s� Crisis 1’� I Ssv#{149}retoxicity

    ‘ No activity

    Programmeddrug death

    (pharmacoptosis)

    inhibits the synthesis of polyamines, was an attractive agent to

    try against the disease. In addition, mitoguazone was interesting

    to try because: (a) the agent already had documented activity in

    patients with non-AIDS-related non-Hodgkin’s lymphoma (12,

    13); (b) the agent causes minimal myelosuppression and other

    toxicities on the new schedule (12, 13); and (c) mitoguazone

    crosses the blood brain barrier, as evidenced by high concen-

    trations in brain tumors (22).

    Recently, Levine et a!. (23) reported the results of a Phase

    II trial of mitoguazone given at a dose of 600 mg/m2 on days I,8, and then every 2 weeks, in patients with AIDS-related lym-phoma. In that study, she treated 35 patients with AIDS-related

    lymphoma, all of whom had failed on prior regimen and 17 of

    whom had failed 2-6 prior regimens. Twenty-four (80%) of the

    patients had a CD4 count of < lOO/dl. There were three patients

    who attained a complete response, plus three who attained a

    partial response for an overall response rate of 6 of 35 or 17%

    (95% confidence interval, 4.7-29.6%; Ref. 23). A second study

    in a similar population has confirmed that response rate (24).

    The toxicities associated with the use of mitoguazone on the

    days 1 , 8, and then every 2 weeks schedule were minimal, with

    only an 8% incidence of grades 3 and 4 mucositis. The lesson

    learned from the further development of mitoguazone is that

    new knowledge regarding the basis for toxicity with a corn-

    pound (in this case, its clinical pharmacology) can lead to adifferent way of using that compound that avoids the toxicities

    and allows its antitumor activities to be exploited.

    After the presentation of this lectureship, the Phase II dataon the activity of mitoguazone in patients with AIDS-related

    non-Hodgkin’s lymphorna was presented to the Food and Drug

    Administration Advisory Committee, and the Committee rec-

    ommended against its approval based only on Phase II data. Arandomized Phase HI trial to further support its efficacy wasrecommended by the Food and Drug Administration, and plans

    are being made to conduct that study. The drug has not yet

    emerged from pharmacoptosis.

    Why Compounds Don’t Make It-Lack of Efficacy

    A second reason new agents don’t make it into routine

    clinical use is no efficacy (or a supposed lack of efficacy). Many

    NH2

    \/\ /\H2N\,NV N II N NH2 2HCI . H20

    H2N

    0 5 10 15 20

    Days

    investigators would argue that the reason we have so many

    compounds that make it into clinical trials that have poor to no

    activity in the clinic is the poor (nonpredictive) preclinical

    systems we have for selecting those agents (25, 26). Our thesis

    in San Antonio is that in our clinical trial designs, we are asking

    the drug to do more clinically than it did preclinically. For

    example, take the hypothetical new drug “difungomuctane” (nota real drug), which inhibits tumor growth to the degree noted in

    Fig. 4. As can be seen in Fig. 4, difungomuctane clearly causes

    tumor growth delay. However, can we really expect this drug to

    shrink tumors in patients when all it does in animal systems is

    slow down tumor growth? Again, the theory is that with our

    current clinical trial designs, we are asking our drugs to do more

    clinically than they do in preclinical systems. Perhaps new

    clinical trial end points besides shrinkage of patients’ tumors are

    needed to assess the antitumor activity of some new agents.

    An Example of Recovery from Pharmacoptosis Becauseof Supposed Lack of Antitumor Activity. Gemcitabine (LY

    18801 1, difluorodeoxycytidine; dFdC) is a nucleoside with the

    structure shown in Fig. 5. The compound was synthesized by

    Hertel et a!. (27) with tumor growth delay documented in

    several animal models and human tumor xenograft models

    including X5563 myeloma; 6C3HED lymphosarcoma; Ll 210,

    P388, and P1543J leukemia; and the CX-l and LX-1 xenografts

    (28 -30). In addition, the agent had a broad spectrum of antitu-

    mor activity against human tumor colony-forming units taken

    directly from patients and growing in soft agar (31). Activity

    was particularly impressive against breast, non-small cell lung,

    ovarian, and pancreatic cancer colony-forming units.

    Phase I trials were conducted with gemcitabine beginning

    Research. on June 13, 2021. © 1998 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • < Gemcitabine(1000 mg/m2/week)

    5.Fluorouracil(600 mg/m2/week)

    F

    Fig. 5 Structure of gemcitabine.

    1082 There Are No Bad Anticancer Drugs

    ‘ The abbreviations used are: ‘fl’P, time to tumor progression; DFMO,difluoromethylornithine (eflornithine); ODC, ornithine decarboxylase.

    in 1987 (32, 33). The daily X 5, 30-mm infusion schedule gave

    a maximally tolerated dose of only 9 mg/rn2, with hypertension

    and flu-like symptoms being the dose-limiting toxicities. On the

    every 2 weeks schedule, over 0.5-4 h, the maximally tolerated

    dose was 3600 mg/rn2. Dose-limiting toxicities included neu-

    tropenia and flu-like symptoms. In that study, we noted one

    patient with pancreatic cancer with clinical improvements in-

    cluding less pain, an improved performance status, and weight

    gain. Based on that observation as well as observations from

    other groups, a Phase II trial of gemcitabine was conducted by

    Casper et a!. (34, 35). In that study, 43 patients with advanced

    pancreatic cancer with no prior chemotherapy received doses of

    gemcitabine ranging from 800-1250 mg/m2 weekly X 3 re-

    peated every 28 days. Five partial responses were noted for an

    overall response rate of 13%. Toxicities were mild. What was

    striking was that, despite the overall low incidence of partial

    response (only 13%), there was a far greater percentage of

    patients who reported improvements in their pancreatic cancer-

    related symptoms, i.e., pain, weakness, and weight loss. Because

    virtually no chemotherapy has been shown to impact the symp-

    torns of pancreatic cancer, it was decided to design a clinical

    trial with gemcitabine to determine, in a controlled setting,

    whether that agent decreased the symptoms associated with

    advanced pancreatic cancer. This thinking was a departure from

    the usual oncology clinical trial designs, where typical end

    points are either tumor shrinkage (complete or partial responses)

    or survival. However, in other areas of drug development, other

    end points are always used in clinical trials. For example, the

    end point for a clinical trial for a new antiarrhythmic agent is

    control of arrhythmias, not survival. The end point for the

    clinical trials of a new antihypertensive is control of blood

    pressure, not survival. New end points in clinical trials in

    patients with advanced cancer may also be in order. One of

    those end points could be whether the new therapy improves the

    Patients with advanced,symptomatic pancreaticcancer

    - No prior chemotherapy

    - Pain, intensity score of � 20

    (out of 100 on MPAC*). Analgesic consumption

    � 10 morphine sulfateequivalents

    . Karnofsky PerformanceStatus � SO but � 80

    *MPAC = Memorial Pain Assessment Card

    Fig. 6 Design for definitive trial of gemcitabine in patients with ad-

    vanced symptomatic pancreatic cancer.

    tumor-induced problems that bother the patient. In the case of

    pancreatic cancer, the things that bother the patient include pain,

    weakness, and a declining performance status and weight loss

    (36, 37). If a new treatment could improve those symptoms for

    a patient, we postulated that the new treatment would give them

    clinical benefit and should be made available to them.

    To determine whether gemcitabine gave clinical benefit to

    patients with advanced pancreatic cancer, the clinical trial out-

    lined in Fig. 6 was designed. As can be seen in that figure, to be

    eligible for the study, patients had to have advanced pancreatic

    cancer which was symptomatic (e.g., patients with pain of >20

    of 100 on the Memorial Pain Assessment Card (38); patients

    required more than 10 mg of morphine sulfate or equivalent to

    control their pain; and patients had to have a Karnofsky Per-

    formance Status of >50 but

  • Table 2 Summary of results of randomized trial of gemcitabineversus 5-fluorouracil (5-FU) for treatment of patients with advanced

    pancreatic cancer”

    Fig. 7 Preclinical-clinical uncoupling in drug development.

    Clinical Cancer Research 1083

    Parameter 5-FU arm Gemcitabine arm P

    Clinical benefit 4.8% 23.8% 0.0022Response rate 0% 5.4% 0.077TTP (mo) 1.0 3.2 0.0002Survival

    Median (mo) 4.41 5.65 0.0025One year 2%

    “Bums et al. (42).

    18%

    had clinical benefit versus 5% of the patients who received

    5-fluorouracil (P 0.0022). This has now been reported in

    other trials (43, 44). The TTP (which corresponds best to the

    tumor growth delay in animals) was also significantly better. Of

    additional note and importance is that median survival and

    1-year survival were significantly better for the gemcitabine-

    treated patients. This finding helped validate the use of our

    clinical benefit parameter as a clinically meaningful end point.

    Also importantly, the side-effects profile of gemcitabine was not

    significantly different from the profile of 5-fluorouracil, except

    that more transfusions were required in the patients receiving

    gemcitabine, and neutropenia was also more common. How-

    ever, the gemcitabine patients also lived longer to obtain those

    transfusions or to develop neutropenia.

    The gemcitabine example is provided to document that if a

    new agent produces tumor growth delay in a preclinical model,

    we should strive to use a tumor growth delay end point (e.g.,

    ‘FTP) in our clinical trials. Above all, we should strive to match

    our clinical trial end points to what has been found in preclinical

    models. Only then will there be a true and fair clinical test for

    that new agent. This will be particularly important as our new

    molecular biology gives us new agents that are growth-modu-

    lating agents rather than cytotoxic agents that shrink established

    tumors (see below).

    Why Compounds Don’t Make It-Attention Is Not

    Paid to the Mechanisms of Action of the Compound

    When attention is not paid to the mechanism of action of a

    new compound, our clinical trials often do not exploit that

    mechanism of action, which usually leads to failure of the

    compound in the clinic. As noted in Fig. 7, I refer to this as

    “preclinical-clinical uncoupling.” Table 3 gives some examples

    of preclinical-clinical uncoupling. As can be seen in that table,

    regardless of the mechanism of action of the agent, the only

    parameter we have used to determine whether to continue de-

    velopment of a new agent has been to assess whether the

    compound causes complete tumor shrinkage (100% disappear-

    ance of the tumor lasting at least 1 month) or a partial response

    (�50% disappearance of tumor lasting at least 1 month). This

    nearly total neglect of the mechanism of action probably has led

    to discontinuing the development of many new agents that, had

    their mechanism of action been remembered and our trials

    designed to measure that mechanism, may have been successful

    in the clinic.

    An Example of an Agent Recovering from Pharmacop-tosis Because of Attention Paid to Its Mechanism of Action.

    DFMO has the structure shown in Fig. 8. The compound has a

    unique mechanism of action, i.e., the irreversible inhibition of

    ODC (45, 46). ODC is becoming a more interesting target in

    both the areas of cancer treatment and in cancer etiology and,

    hence, in cancer prevention. More specifically, ODC has been

    found to be up-regulated in the tissues of several premalignant

    conditions, including cervical intraepithelial neoplasia (47), co-

    lon polyps (48), and others (49). Manni et a!. (50) have found

    that ODC is up-regulated in patient breast cancer tissue, and

    elevation of the expression of ODC in that tissue is of greater

    significance for a poor prognosis than is the estrogen receptor

    status or the number of positive lymph nodes. Finally, of great

    interest, is that Auvinen et al. (51) have reported that ODC is an

    oncogene. Transfection of the ODC gene into NIH 3T3 cells

    causes transformation of the cells.

    As noted above, DFMO is an irreversible inhibitor of ODC.

    DFMO blocks the promotion step of carcinogenesis in several

    animal models including breast, colon, bladder, skin, and pros-

    tate models (52-55). Unfortunately, the initial clinical trials with

    DFMO developed the agent as a standard anticancer agent with

    escalation to the maximally tolerated dose of the agent. Toxic-

    ities noted with doses of �20 g/m2/day of DFMO included

    thrombocytopenia, nausea and vomiting, leukopenia, anemia,

    diarrhea, high frequency hearing loss, and in some cases, met-

    abolic acidosis (56, 57). After those studies, DFMO was aban-

    doned (underwent pharmacoptosis) by most investigators.

    A breakthrough in the use of DFMO was a study by Love

    et al. (58) who performed a clinical trial to attempt to exploit the

    mechanism of action of DFMO, the inhibition of ornithine

    decarboxylase. They conducted a study to determine the small-

    est dose of DFMO that could be used to inhibit ornithine

    decarboxylase by carefully performing skin biopsies on the

    patients. They found that a dose of 0.5 g/m2/day (one-twentieth

    the daily dose used in prior cytotoxic approach studies) inhibited

    ornithine decarboxylase. Also, importantly, they noted that no

    toxicity was seen at that dose level. This finding fostered addi-

    tional clinical trials because DFMO could now be used as a

    chemoprevention agent.

    The field of chemoprevention has changed substantially

    over the past few years because there are now histological

    changes that are regarded as premalignant. These premalignant

    changes include cervical intraepithelial neoplasia (CIN III),

    actimc keratosis, colon adenoma, ductal carcinoma in situ of the

    breast, previously resected superficial bladder cancer, atypical

    squamous metaplasia/dysplasia in the lung, oral dysplastic leu-

    koplakia, and prostatic intraepithelial neoplasia. The goal in

    chemoprevention now is to reverse those premalignant changes

    Research. on June 13, 2021. © 1998 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • CHF2

    H2N - CH2 - CH2 - CH2- C - COOH

    NH2

    Fig. 8 Structure of DFMO.

    Period A

    I (Standard �Therapy)

    Period B

    (New Agent) �1

    - Ratio Period B = Growth Modulation IndexPeriod A-Anything > 1.33 (33% improvement) is

    considered excellent (and is unexpected forsecond-line therapy)

    Fig. 9 A method for early determination as to whether a new agent ishaving a modulating effect on tumor growth.

    /7� Standard agent + Placebo

    Standard agent + new agent

    1084 There Are No Bad Anticancer Drugs

    with end points of ‘FTP and 1-year survival.

    Table 3 Examples of preclinical-clinical uncoupling

    What is seen preclinically What we are measuring clinically

    1. The agent inhibits angiogenesis Partial or complete tumor shrinkage

    2.

    3

    The agent prevents metastases

    The agent induces apoptosis

    Partial or complete tumor shrinkage

    Partial or complete tumor shrinkage

    (use them as an intermediate marker for an effect of the agent

    being studied; Ref. 59).

    To date, three clinical trials have correctly used low doses

    of DFMO to inhibit ODC. Mitchell et a!. (47) have recently

    published their work demonstrating that DFMO could substan-

    tially reduce (in 52% of patients) cervical intraepithelial neo-

    plasia (CIN-Ill, a substantial risk factor for the development of

    cervical cancer) after only one month of use. Meyskens et al.

    (60) have clearly documented that low doses of DFMO can

    suppress polyamine content in colorectal biopsy specimens as

    preparation for their randomized trial of a nonsteroidal, anti-

    inflammatory agent plus DFMO versus the nonsteroidal agent

    alone in patients with a history of recurrent colorectal polyps.

    The end point for this study will be the number of polyps that

    recur per unit of time.

    Last, but not least, is a trial published by Alberts et a!. (61),

    who randomized 44 patients with actinic keratoses (a premalig-

    nant skin condition) to receive either DFMO cream or a placebo

    cream. This randomized, double-blinded trial showed a signif-

    icant decrease in actinic keratoses with three months of use of

    the DFMO cream, whereas there was no decrease with the

    placebo cream (61). There was a reduced spermidine:spermine

    ratio in the skin biopsies from the patients receiving DFMO,

    indicating that DFMO was inhibiting the ODC. This is a proof

    of principle that inhibition of the ODC by DFMO resulted in a

    clinically measurable decrease in the number of actinic kerato-

    ses after only a short period of application.

    How Can We Measure a Clinical Effect of a New

    Compound That Is Not a Cytotoxic Agent?

    The developments in molecular biology and the identi-

    fication of new targets provide challenges to both the bench

    and the clinical investigator (62). There are new antisense

    molecules, farnesyl transferase inhibitors, telomerase inhib-

    itors, kinase inhibitors, angiogenesis inhibitors, and many

    other molecules that are likely to be growth-modulating and

    that will not cause immediate tumor shrinkage. One way to

    test a growth-modulating agent is to conduct a randomized

    trial, as was done for gemcitabine, with end points of time-

    to-tumor progression and 1-year survival. However, is there

    a way we can tell early in its development whether an agent

    has any promise (before conducting the randomized Phase III

    trial)? One way that might be used to obtain an early lead as

    to whether a growth-modulating agent is having an effect is

    outlined in Fig. 9. Our hypothesis is that if the new agent has

    an antitumor effect, it will change the natural history of the

    disease. Therefore, we will use the patient as his/her own

    control. As can be seen in Fig. 9, period B is the TTP on the

    new agent. Period A is the TTP on the treatment the patients

    received just before the new agent was started. Given the

    natural history of cancer, it would be shorter that the TTP on

    the new agent would be shorter than the TTP on a prior

    regimen. Therefore, if the TTP on the new agent (period B)

    is greater than the TTP on the prior therapy (period A) it is

    likely the new agent is having an effect on the natural history

    of that patient’s tumor. In Fig. 9, we suggested a 33%

    improvement; however, that value is arbitrary. Although not

    yet tested prospectively, the use of a patient as his/her control

    may be a way to determine whether a growth-modulating

    agent is having a clinical effect before the randomized Phase

    III trial is launched.

    Summary of Lessons Learned

    What lessons have we learned to increase the percentage of

    our agents that make it in the clinic? The first lesson is to avoid

    pharmacoptosis (programmed drug death). If the new agent

    shows toxicities in the clinic but has hints of antitumor activity,

    be persistent and find a way to circumvent that toxicity.

    The second lesson is for the correct clinical trial design to

    mimic the preclinical activity of the agent. For a growth-mod-

    ulatory agent, a suggested clinical trial design would be:

    Research. on June 13, 2021. © 1998 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • Clinical Cancer Research 1085

    The third lesson is to take advantage of the mechanism of

    action of the new agent when designing a clinical trial. For an

    angiogenesis inhibitor, use a minimal residual disease model

    such as:

    . . Angiogenesis inhibitor

    put patient in bestremission (minimal

    residual disease) Placebo

    with end points of ‘FTP or recurrence-free survival.

    The fourth lesson (for the preclinical scientist) is to demand

    better trial designs for your molecule! The fifth lesson for my

    clinical colleagues and myself is to take it upon ourselves to

    personally give new agents the best mechanism-of-action-based

    trial design we possibly can.

    Conclusion

    In conclusion, I would like to thank all of the patients,

    mentors, and colleagues who have made this work possible. San

    Antonio is the birthplace of Texas freedom, and we always say,

    “Remember the Alamo!” From this lectureship, I would like you

    to remember that both bench investigations and clinical inves-

    tigations are difficult endeavors and that we need to appreciate

    both of them as important sciences. Remembering that will keep

    our clinical trial designs optimal and provide the best clinical

    development of a new idea or a new agent from the bench.

    Acknowledgments

    I gratefully acknowledge the expert assistance of Laida Garcia,Meredith Sterling, and Judy Turner in preparing the manuscript for

    publication.

    References

    1. Von Hoff, D. D. MGBG: teaching an old drug new tricks. Ann.

    Oncol., 5: 487-493, 1994.

    2. Thiele, J., and Dralle, F. Zur Kenntnis des amino gunidins. I. Con-densations-produkte de amidoguanidins mit aldehyden and ketonen derfettreihe. Justus Liebigs Ann. Chem., 302: 275-299, 1898.

    3. Corti, A., Dave, C., Williamson-Ashman, H. G., Mihich, E., and

    Schenone, A. Specific inhibition of the enzymatic decarboxylation ofS-adenosylmethionine by methylglyoxal bis(guanylhydrazone) and re-lated substances. Biochem. J., 139: 351-357, 1974.

    4. Pleshkewych, A., Kramer, D. L., Kelly, E., and Porter, C. W. Inde-

    pendence of drug action on mitochondria and polyamines in L1210

    leukemia cells treated with methylglyoxal bis(guanylhydrazone). Can-

    cer Res., 40: 4533-4540, 1980.

    5. Pine, M. J., and DiPaolo, J. The antimitochondrial actions of2-chloro-4’,4”-bis(2-imidazolin 2-yl)-terephthalanilide and methyl-

    glyoxal bis(guanylhydrazone). Cancer Res., 26: 18-25, 1966.

    6. Freireich, E., Frei, E., III, and Karon, M. Methylglyoxal bis(guanyl-hydrazone): a new agent active against acute myelocytic leukemia.Cancer Chemother. Rep., 16: 183-186, 1962.

    7. Regelson, W., and Holland, J. F. Clinical experience with methyl-

    glyoxal bis(guanylhydrazone) dihydrochloride: a new agent with clini-cal activity in acute myelocytic leukemia and the lymphomas. CancerChemother. Rep., 27: 15-26, 1963.

    8. Levin, R. H., Henderson, E., Karon, M., and Freireich, E. J. Treat-

    ment of acute leukemia with methylglyoxal bis(guanylhydrazone)

    (methyl-GAG). Clin. Pharmacol. Ther., 6: 31-42, 1964.

    9. Marsh, K. C., Liesmann, J., Patton, T. F., Fabian, C. J., and Stemson,L. A. Plasma levels and urinary excretion of methyl-GAG following iv.infusion in man. Cancer Treat. Rep., 65: 253-257, 1981.

    10. Hart, R. D., Roboz, J., Wu, K., Bruckner, T., Ohnuma, T., andHolland, J. F. Clinical and pharmacologic studies with weekly andbiweekly methylglyoxal bis-guanylhydrazone (methyl-G). Proc. Am.

    Assoc. Cancer Res., 21: 181, 1980.

    11. Rizzo, J., Levine, A. M., Weiss, G. R., Pearce, T., Kraynak, M.,

    Mueck, R., Smith, S., Von Hoff, D. D., and Kuhn, J. G. Pharmacoki-netics profile of mitoguazone (MGBG) in patients with AIDS-related

    non-Hodgkin’s lymphoma. Invest. New Drugs, 14: 227-234, 1996.

    12. Knight, W. A., III, Fabian, C., Costanzi, J. J., Jones, S. E., and

    Coltman, C. A., Jr. Methylglyoxal bis-guanylhydrazone (methyl GAG,MGBG) in lymphoma and Hodgkin’s disease. Invest. New Drugs, 1:

    225-237, 1983.

    13. Warrell, R. P., Lee, B. J., Kempin, S. J., Lacher, M. J., Straus, D. J.,

    and Young, C. W. Effectiveness of methyl-GAG (methylglyoxal-bis-[guanylhydrazone]) in patients with advanced malignant lymphoma.Blood,57: 1011-1014, 1981.

    14. Warrell, R. P., and Burchenal, J. H. Methylglyoxal- bis(guanylhy-

    drazone) (methyl GAG): current studies and future prospects. J. Clin.

    Oncol., 1: 52-65, 1983.

    15. Luedke, D. W., Maddox, W., Birch, R., Velez-Garcia, E., andSchleuter, J. Mitoguazone in advanced squamous cell carcinoma of head

    and neck origin: a Phase II trial of the Southeastern Cancer Study Group.Cancer Treat. Rep., 70: 529-530, 1986.

    16. Scher, H. I., Yagoda, A., Ahmed, T., and Watson, R. C. Methyl-glyoxal-bis(guanylhydrazone) in hormone-resistant adenocarcinoma of

    the prostate. J. Clin. Oncol., 3: 224-228, 1985.

    17. Karp, J. E., and Broders, S. Acquired immunodeficiency syndrome

    and non-Hodgkin’s lymphoma. Cancer Res., 51: 4743-4756, 1991.

    18. Levine, A. Acquired immunodeficiency syndrome related lym-

    phoma. Blood, 80: 8-20, 1992.

    19. Kaplan, L. D., Straus, D. J., Testa, M. A., Van Roenn, J., Dezube,B. J., Cooley, T. P., Hemdier, B., Northfelt, D. W., Huang, J., Tulpule,

    A., and Levine, A. M. Low-dose compared with standard-dosemBACOD chemotherapy for non-Hodgkin’s lymphoma associated with

    human immunodeficiency virus infection. N. Engl. J. Med., 336: 1641-1648, 1997.

    20. Russell, D. H., Levy, C. C., Schimpft, S. C., and Hawk, I. A.

    Urinary polyamine in cancer patients. Cancer Res., 31: 1555-1558,

    1971.

    21. Russell, D. H. Increased polyamine concentrations in the urine of

    human cancer patients. Nature (Lond.), 233: 144-145, 1971.

    22. Rosenblum, M. G., Stewart, D. J., Yap, B. S., Leavens, M., Ben-

    jamin, R. S., and Loo, T. L. Penetration of methylglyoxal bis(guanyl-hydrazone) into intracerebral tumors in humans. Cancer Res., 41: 459-

    462, 1981.

    23. Levine, A. M., Tulpule, A., Tessman, D., Kaplan, L., Giles, F.,

    Luskey, B. D., Scadden, D. T., Northfelt, D., Silverberg, I., Wernz, J.,Espina, B., and Von Hoff, D. D. Mitoguazone therapy in patients with

    refractory or relapsed AIDS-related lymphoma: results from a multi-

    cancer Phase II trial. J. Clin. Oncol., 15: 1094-1 103, 1997.

    24. Levine, A. M., Giles, F., Kaplan, L, Sparano, J., Scadden, D.,

    Luskey, B., Northfelt, D., Thommas, J., Anderson, L. J. J., Espina, B.,Tessman, D., and Von Hoff, D. Two sequential prospective, multi-institutional Phase II clinical trials of mitoguazone (MGBG) in refrac-tory or relapsed AIDS-related lymphoma. Blood, 86 (Suppl 1): 381a,1995.

    25. Staquet, M. J., Byar, D. P., Green, S. P., and Rozenwey, M. Clinicalproductivity of transplantable tumor system in the selection of new

    drugs for solid tumors: rationale for a three-stage strategy. Cancer Treat.

    Rep., 67: 753-765, 1983.

    26. Twentyman, P. Drug screening in xenografts: the European multi-

    center project. Ann. Oncol., 5: 394-396, 1994.

    27. Hertel, L. W., Boder, G. B., Kroin, J. S., Rinzei, S. M., Poore, G. A.,

    Todd, G. C., and Grindey, G. B. Evaluation of the antitumor activity ofgemcitabine (2’,2’-difluoro-2’-deoxycytidine). Cancer Res., 50: 4417-4422, 1990.

    Research. on June 13, 2021. © 1998 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • 1086 There Are No Bad Anticancer Drugs

    28. Heinemann, V., Hertel, L., Grindey, G. B., and Plunkett. W. Com-

    parison of the cellular pharmacokinetics and toxicity of 2’,2’-difluoro-deoxycytidine and 1 .43-D-arabinofuranosyl cytosine. Cancer Res., 48:

    4024-4031, 1988.

    29. Ghandi, V., and Piunkett, W. Modulatory activity of 2’,2’-difluoro-

    deoxycytidine on the phosphorylation and cytotoxicity of arabinosyl

    nucleosides. Cancer Res., 50: 3675-3680, 1990.

    30. Huang. P., Chubb, S., Hertel, L. W., Gnndey, G. B., and Plunkett,W. Action of 2’,2’-difluorodeoxycytidine on DNA synthesis. Cancer

    Res.,5/:6l10-6l17, 1991.

    31. Hanauske, A., Degen, D., Marshall, M. H., Hilsenbeck, S. G.,

    Grindey, G. B., and Von Hoff, D. D. Activity of 2’,2’-difluorodeoxy-

    cytidine (gemcitabine) against human tumor colony forming units. An-

    ticancer Drugs, 3: 143-146, 1992.

    32. O’Rourke, T. J,. Brown, T. D., Havlin, K., Kuhn, J. G., Craig, J. B.,

    Burns, H. A., Satterlee, P. G., Tarassoff, F., and Von Hoff, D. D. Phase

    I clinical trial of gemcitabine given as an intravenous bolus on 5

    consecutive days. Eur. J. Cancer, 30A: 417-418, 1994.

    33. Brown, T., O’Rourke, T., Bums, H., Kuhn, J., Tarassoff, P., Cag-

    nola, J., Rodriguez, G., and Von Hoff, D. A Phase I trial of gemcitabine

    (LY18801 1) administered intravenously every two weeks. Proc. Am.Soc. Clin. Oncol. Annu. Meet., 10: 115, 1991.

    34. Casper, E. S., Green, M. R., Brown, T. D., Keisen, D. P., Kresek, T.,

    Trochanowski, B., and Tarassoff, P. G. Phase II trial of gemcitabine

    (2’.2’-difluorodeoxycytidine) in patients with pancreatic cancer. Proc.

    Am. Soc. Clin. Oncol. Annu. Meet., 10: 143, 1991.

    35. Casper, E. S., Green, M. R., Kelsen, D. P., Heelan, R. T., Brown,T. D., Flombaum, C. D., Trochanowski, B., and Tarassoff, P. G. PhaseII trial of gemcitabine (2’,2’-difluorodeoxycytidine) in patients with

    adenocarcinoma of the pancreas. Invest. New Drugs, 12: 29-34, 1994.

    36. Warshaw, A. L., and Fernandez-del Castillo, C. Pancreatic carci-

    noma. N. Engi. J. Med., 326: 455-465, 1992.

    37. Kelsen, D. P., Portenoy, R. K., Thaler, H. T., Niedzwiecki, D.,

    Passik, S. D., Tao, Y., Banks, W., Brennan, M. F., and Foley, F. M. Pain

    and depression in patients with newly diagnosed pancreas cancer.J. Clin. Oncol.. 13: 748-755, 1995.

    38. Fishman, B., Pastemak, S., and Wallenstein, S. L. The Memorial

    Pain Assessment Card: a valid instrument for the evaluation of cancerpain. Cancer (Phila.), 60: 1 151-1 158, 1987.

    39. Andersen, J. S., Burns, H. A., Casper, E., Clayman, M., Green, M.,

    Nelson, R. L., Portenoy, R., Rothenberg, M., Tarassoff, P. G., and Von

    Hoff, D. D. Development of a new system for assessing clinical benefitfor patients with advanced pancreatic cancer. Proc. Am. Soc. Clin.

    Oncol. Annu. Meet., 13: 461, 1994.

    40. Green, S., and Weiss, G. R. Southwest Oncology Group standard

    response criteria, endpoint definitions, and toxicity criteria. Invest. New

    Drugs, 10: 239-253, 1992.

    41. Moore, M., Andersen, J., Burns, H., Tarassoff, P., Green, M.,

    Casper, E., Portenoy, R., Modiano, M., Cripps, C., Nelson, R., Stor-niolo, A., and Von Hoff, D. D. A randomized trial of gemcitabine

    (GEM) versus 5FU as first-line therapy in advanced pancreatic cancer.Proc. Am. Soc. Ciin. Oncol. Annu. Meet., 14: 199, 1995.

    42. Burns, H. A., III, Moore, M. J., Andersen, J., Green, M. R.,

    Rothenberg, M. L., Modiano, M. R., Cripps, M. C., Portenoy. R. K.,

    Storniolo, A. M., Tarassoff, P., Nelson, R., and Von Hoff, D. D.

    Improvements in survival and clinical benefit with gemcitabine as

    first-line therapy for patients with advanced pancreas cancer: a random-

    ized trial. J. Clin. Oncol., 15: 2403-2413, 1997.

    43. Rothenberg, M. L., Moore, M. J., Cripps, M. C., Andersen, J. S.,

    Portenoy, R. K., Burns, H. A., III, Green, M. R., Tarassoff, P. G.,

    Brown, T. D., Casper, E. S., Stomiolo, A-M., and Von Hoff, D. D. A

    Phase II trial of gemcitabine in patients with 5-Hi-refractory pancreas

    cancer. Ann. Oncol., 7: 347-353, 1996.

    44. Carmichael, J., Fink, U., Russell, R. C., Spittle, M. F., Harris, A.,Spiessel, G., and Blatter, J. Phase II study of gemcitabine in patients

    with advanced pancreatic cancer. Br. J. Cancer, 73: 101-105, 1995.

    45. Bey, P.. Danzin, C., Von Dorsselaer, V., Mamont, P., Jung, M., andTardif, C. Analogues of ornithine as inhibitors of omithine decarboxy-

    lase: new deductions conceming the topography of the enzyme’s active

    site. J. Med. Chem., 21: 50-55, 1978.

    46. Pegg, A. E. Polyamine metabolism and its importance in neoplastic

    growth and as a target for chemotherapy. Cancer Res., 48: 759-784,

    1988.

    47. Mitchell, M. F., Tortolero-Luna, G., Lee, J. J., Hittelman, W. N.,

    Lotan, R., Wharton, J. T., Hong, W. K., and Nishioka, K. Phase Idose-escalation trial of a-difluoromethylomithine in patients with cer-

    vical intraepithelial neoplasia grade 3. Clin. Cancer Res., 4: 303-3 10,

    1998.

    48. Porter, C., Herrera-Omelas, L., Pera, P., Retrelli, N. F., and Mittel-

    man, A. Polyamine biosynthetic activity in normal and neoplastic hu-man colorectal tissue. Cancer (Phila.), 60: 1275-1281, 1987.

    49. Garewal, H. S., Sampliner, R., Gemer, E., Steinbronn, K., Alberts,D., and Kendall, D. Omithine decarboxylase activity in Barrett’s esoph-

    agus: a potential marker for dysplasia. Gastroenterology, 94: 8 19-821,

    1988.

    50. Manni, A., Mauger, D., Gimotty, P., and Badges, B. Prognostic

    influence on survival of increased omithine decarboxylase activity in

    human breast cancer. Clin. Cancer Res., 2: 1401-1406, 1996.

    51. Auvinen, M., Paasinen, A., Anderson, L. C., and Holtta, E. Omi-

    thine decarboxylase activity is critical for cell transformation. Nature

    (Lond.), 360: 355-358, 1992.

    52. Thompson, H. J., Meeker, L. D., Herbst, E. J., Ronan, A. M., and

    Minocha, R. Effect of concentration of DL-a-difluoromethylomithine onmurine mammary carcinogenesis. Cancer Res., 45: 1170-1173, 1985.

    53. Reddy, B. S., Nayini, J., Tokumo, K., Rigotty, J., Zang, E., and

    Kelloff, G. Chemoprevention of colon carcinogenesis by concurrentadministration of piroxicam, a nonsteroidal anti-inflammatory drug with

    DL-a-difluoromethylornithine on ornithine decarboxylase inhibitor.

    Cancer Res., 50: 2562-2568, 1990.

    54. Homma, Y., Kakizoe, T., Samma, S., and Oyasu, R. Inhibition of

    N-butyl-N (hydroxybutyl) nitrosamine-induced rat urinary bladder

    carcinogenesis by a-difluoromethylornithine. Cancer Res., 47: 6176-

    6179, 1987.

    55. Weeks, C. E., Herrimann, A. L., Nelson, F. R.. and Sloga. T. J.Difluoromethylomithine, an irreversible inhibitor of ornithine decarbox-

    ylase, inhibits tumor promoter-induced polyamine accumulation and

    carcinogenesis in the mouse skin. Proc. NatI. Acad. Sci. USA, 74:

    6028-6032, 1982.

    56. Abeloff, M. D., Slavik, M., Luk, G. D., Griffen, C. A., Hermann, J.,

    Blanc, 0.. Sjoerdsman, A., and Baylin, S. B. Phase I and pharmacoki-netic studies of a-difluoromethylornithine-an inhibitor of polyamine

    biosynthesis. J. Clin. Oncol., 2: 124-130, 1984.

    57. Griffin, D. A., Slavik, M., Chien, S. C., Hermann, J., Thompson, G.,

    Blanc, 0., Luk, G. D., Baylin, S. B., and Abeloff, M. D. Phase I trial andpharmacokinetic study of intravenous and oral a-difluoromethylomi-thine. Invest. New Drugs, 5: 177-186, 1987.

    58. Love, R. R., Carbone, P. P., Vermia, A. K., Gilmore, D., Carey, P.,

    Tutsch, K. D., Pomplun, M., and Wilding, G. Randomized Phase I

    chemoprevention dose-seeking study of a-difluoromethylornithine.

    J. Natl. Cancer Inst., 85: 732-737, 1993.

    59. Kelloff, G. J., Boone, C. W., Crowell, J. A., Steele, V. E., Lubet, R.,

    and Doody, L. A. Surrogate endpoint biomarkers for Phase II cancer

    chemoprevention trials. J. Cell. Biochem., 19: 1-9, 1994.

    60. Meyskens, F. L., Jr., Emerson, D. S., Pelot, D., Meshkinpour, H.,Shassetz, L. R., Einspahr, J., Alberts, D. S., and Gemer, E. W. Dose

    de-escalation chemoprevention trial of a-difluoromethylornithine in pa-

    tients with colon polyps. J. Natl. Cancer Inst., 86: 1 122-1130, 1994.

    61. Alberts, D. S., Dorr, R. T., Aikin, M., Quinn, J., Goldman, R.,Nelson, M., Xu, M. J., Peng, Y. M., Bozzo, P., and Gensler, H. Positive

    randomized, double-blinded, placebo-controlled study of topical diflu-

    oromethylomithine (DFMO) in the chemoprevention of skin cancer.

    Proc. Am. Soc. Clin. Oncol. Annu. Meet., 15: 172, 1996.

    62. Schipper, H., Goh, C. R., and Wang, T. L. Shifting the cancer

    paradigm: must we kill to cure? J. Clin. Oncol., 13: 801-807, 1994.

    Research. on June 13, 2021. © 1998 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • 1998;4:1079-1086. Clin Cancer Res D D Von Hoff Award Lecture.designs--twenty-first Richard and Hinda Rosenthal Foundation There are no bad anticancer agents, only bad clinical trial

    Updated version

    http://clincancerres.aacrjournals.org/content/4/5/1079

    Access the most recent version of this article at:

    E-mail alerts related to this article or journal.Sign up to receive free email-alerts

    Subscriptions

    Reprints and

    [email protected] at

    To order reprints of this article or to subscribe to the journal, contact the AACR Publications

    Permissions

    Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

    .http://clincancerres.aacrjournals.org/content/4/5/1079To request permission to re-use all or part of this article, use this link

    Research. on June 13, 2021. © 1998 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/content/4/5/1079http://clincancerres.aacrjournals.org/cgi/alertsmailto:[email protected]://clincancerres.aacrjournals.org/content/4/5/1079http://clincancerres.aacrjournals.org/