throbber
Clinical Development of Anticancer A
`Perspective
`
`gents—A National Cancer Institute
`
`Silvia Marsoni* and Robert Wittes1
`
`Since the first report that a chemical could cause sig-
`nificant tumor shrinkage (1), the development of clin-
`ically useful antineoplastic drugs has grown from the
`preoccupation of a few investigators to a major interna-
`tional effort. Over the past 35 years or so, about 30
`drugs have been defined as active in one or more tumor
`types. When used alone in patients with disseminated
`mahgnancy,
`these drugs cause reduction in bulk of
`measurable neoplasm in a significant percent of cases;
`for most tumor types, however, ample evidence of re-
`51dual cancer usually persists and after a few months,
`tumor regrowth occurs. More striking successes have
`been achieved with combinations of drugs; as is well
`known, for several kinds of disseminated human can-
`cers, a high frequency of clinical complete remissions,
`with substantial long-term disease-free survival rates,
`is now possible (2—5). For other cancers which may not
`be curable by chemotherapy once they have disseminat-
`ed, combinations of drugs appear to result in a higher
`total remission rate and a greater prolongation of life
`than smgle drugs (6,7). Perhaps more significant in the
`long run is the apparent effect of chemotherapy when
`(1;
`used as part of a planned multimodality effort
`13y some perverse quirk of fate, chemotherapy seems to
`,
`chiefly exert a major impact in rare tumors, while the
`common
`epithelial
`neoplasms
`of
`adulthood
`have
`thus far resisted satisfactory solutions. Therefore, the
`central problem of drug development, the identification
`of effective agents with reasonable therapeutic index,
`IS as pertinent for oncology now as at any time in the
`past.
`The idealized outlines of the successive steps in drug
`development are familiar to all oncologists. In phase I
`trials, we define a dose suitable for use in studies of
`the drug’s activity across a spectrum of human tumors.
`Increasing awareness of the importance of patient— and
`disease-related parameters has effectively led to the re-
`placement of the broad phase II trial with a series of
`disease-oriented activity studies. Having defined set-
`
`investigators
`tings in which the new drug is active,
`then proceed to compare the new treatment with stand-
`ard therapy (phase III) and to further explore the
`drug’s therapeutic potential in other ways, such as in
`combination with other agents or by alternate routes of
`administration.
`
`Anyone familiar with the actual workings of this
`process over the past two decades knows that despite
`its successes, it has not functioned as systematically or
`efficiently as the above description might imply. In ad-
`dition, many of the assumptions on which the process
`was based are in need of reexamination. Since there
`are
`no
`reliable
`laboratory predictors of
`efficacy
`for specific human cancers, drug development will con-
`tinue to require extensive testing in human subjects, an
`endeavor that is never without ethical dilemmas, how-
`ever thoughtfully it is carried out. In addition, because
`human cancers vary widely in sensitivity to anticancer
`drugs,
`the apparatus required to sustain the clinical
`trials effort is necessarily large and very expensive. For
`these reasons alone, another look at the drug develop-
`ment program of the National Cancer Institute (NCI)
`seems to be worthwhile.
`
`Phase I
`
`Phase I trials of antineoplastic compounds are con-
`ducted in patients with disseminated malignancies for
`whom standard treatment either does not exist or has
`proved ineffective. Drugs are given in a phase I trial
`with therapeutic intent; the main scientific goal is to
`define the qualitative and quantitative characteristics
`of the drug’s acute toxicity, and in so doing, to deter-
`mine a biologically active dose which is tolerable for ev-
`ery patient. The maximum tolerated dose (MTD)
`is
`usually higher in children than in adults (10), probably
`because of better organ function and possibly because
`of different pharmacokinetics (11,12). Also, since the
`MTD for patients with acute leukemia is often substan-
`tially higher than that for solid tumors, at least four
`
`I
`
`.
`.
`.
`.
`Cancer Therapy Evaluation Program, Dmsnon of Cancer Treatment.
`Na'tional Cancer Institute, Bethesda, MD.
`Reprint requests to: Silvia Marsoni, MD, Drug Evaluation and Re
`
`.
`porting Section, Investigational Drug Branch, Cancer Therapy Evaluation
`Program, National Cancer Institute, Landow Bldg, Rm 4C09, 7910 Wood-
`mont Ave, National Institutes of Health, Bethesda, MD 20814.
`
`Cancer Treatment Reports Vol. 68, No. 1, January 1984
`
`77
`
`This mate ria i was copied
`
`Celltrion v. Genentech
`
`IPR2017—01122
`
`Genentech Exhibit 2023
`
`

`

`trials should be conducted for each drug. In
`phase I
`practice, of course, three to four phase I trials testing
`different schedules of the drug are usually performed
`in adult patients with solid tumor alone, and trials in
`children do not start until substantial experience is ac-
`cumulated from the adult trials.
`The assumption underlying all phase I escalation pro-
`cedures is that anticancer compounds must be given at
`or near the MTD; therefore, the job of a phase I trial is
`to define the highest dose that can be safely delivered
`to a patient, since this dose will also be the one that
`has the best chance of being active. This approach re-
`flects the difficulties in establishing a clear-cut measur-
`able endpoint for drug activity against cancer. Since
`induction of response is not usually an event that is im-
`mediately recognizable, attainment of toxicity is the
`only assurance that, if a response is not obtained, at
`least a biologically active dose was delivered. Underly-
`ing this assumption is the more fundamental one that
`the dose-response curve f0r human cancers is monoton-
`ically increasing throughout
`the range of
`tolerated
`doses and is to the right of the dose-toxicity curve.
`Needless to say, the details of this assumption have not
`been generally verified for antitumor agents, chiefly
`because rigorously defining the shape of a clinical dose-
`response curve is a laborious task, requiring a large
`number of patients treated at each of several dose lev-
`els. Where the relationship between dose and response
`has been examined, however, most of the data are at
`least consistent with the conclusion that the higher the
`administered dose, the more probable an antitumor ef-
`fect (13-17) or the longer the duration of remission
`(18). On the other hand, recent trials in small cell lung
`cancer suggest that the probability of response does
`not continue to increase linearly as the dose approaches
`the MTD (19).
`For most of the clinically useful compounds, the bone
`marrow is dose-limiting. The dose-toxicity curve for
`
`myelosuppression is quite reproducible, and the status
`of marrow reserve is the major source of interpatient
`variability. Generally, treatment of six to ten patients
`at or near the MTD is sufficient to establish a safe
`phase II dose when myelosuppression is the dose-limit-
`ing toxicity.
`However, major problems may arise when other toxic
`effects which are less easily quantifiable are dose-limit-
`ing. For example, in a phase I study of escalating doses
`of carmustine with autologous bone marrow support
`(20), major organ toxicity (liver, central nervous system,
`and lung) surfaced abruptly at a dose of 1500 mg/m2.
`Because of
`the sudden appearance of
`these side
`effects in the escalation scheme and the long interval
`from the beginning of treatment to onset of toxicity
`(6-9 weeks), the overall mortality rate for patients en-
`tered at a dose of 2 1500 mg/m2 was approximately
`30%.
`
`Experience suggests that whenever a drug has dose-
`limiting side effects other than myelosuppression, its
`transition into phase II has often been compromised.
`An analysis of 31 drugs entered in phase I evaluation
`by the NCI shows that whenever the drug had myelo-
`suppression alone as the dose-limiting toxicity, it had a
`high probability of undergoing full phase II study; how-
`ever, when other organ toxicity was dose-limiting, only
`25% of the drugs proceeded to full phase II study (ta-
`ble 1). Evaluation of the remainder of drugs was re-
`stricted by either the NCI or lack of investigator inter-
`est. The main reason for these difficulties relates large-
`ly to the uncertainty regarding reversibility of acute
`major organ damage. In addition, even if organ damage
`should turn out to be reversible, medical support dur-
`ing periods of severe organ failure is either extremely
`intensive and costly (kidney, CNS) or technically unsat-
`isfactory (liver), and is not seen as feasible or justifia-
`ble by most investigators in the context of a clinical ex-
`periment.
`
`TABLE 1.——Pliase II evaluation as a function of the dose-limiting toxicity of 31 cytotoxic compounds (1975- 1982)
`
`Dose-limiting toxicitym
`Myelosuppressicn
`Organ
`and organ toxicityi"
`toxicity”:
`3
`3
`3
`4
`
`Myelosuppression *
`10
`
`MF
`
`Phase II
`
`evaluation
`ull
`
`Restricted
`Dropped
`Toxicity
`No interest
`No drug supply
`
`0
`
`0
`1
`1
`
`0
`l
`0
`
`4
`1
`0
`
`12
`Total
`7
`
`'Biszmtrene, (liaziquOne, ziclarubicin (aclacinomycin). mitoxantrone, PCNU, amsacrinc. zorubicin. ChIOI'OZOtociIL
`carboplatin, ICRI“ 187, 5-metliyltetrahydrohomofolate, and 3-(lcnzauridine.
`JrAcivicin, maytansine, anguidine, teroxirone, dihydro-5-azacitidine, indicine N-oxide, and DON.
`iPyrazofurin, L-alanosinc, homoharringtonine, 'l‘CN-I’, N-methylformamide, pentostatin, liycantbone, dichloroul-
`lyl lawsone, pyrazole, aminotbiudiazole, bruceantin, and spirogermanium.
`
`12
`
`78
`
`This material was copied
`_xx_a__ LII ll___l _,_r_1_-
`
`Cancer Treatment Reports
`
`

`

`This dilemma appears to have no easy solutions. One
`alternative might be to utilize data from pharmacologic
`studies to define the relationship between dose, plasma
`level, tissue level, and clinical activity. For example, in
`the case of pentostatin, knowledge concerning the
`amount of drug needed to abolish activity of the target
`enzyme adenosine deaminase has helped to establish a
`phase II dose independent of the attainment of clinical
`toxicity. Unfortunately, however, this is an exceptional
`situation and, under most circumstances, specific intra-
`cellular targets of drug action either have not been
`identified or are not so susceptible to analysis.
`A somewhat more empiric approach is exemplified by
`the current plans for developing N-methylformamide, a
`drug which had been introduced into the clinic in the
`19505 and was subsequently dropped while in phase I
`because of hepatotoxicity (21). Interest in the drug has
`recently been revived because of its activity against hu-
`man tumor xenografts (22) and its capacity to induce
`differentiation in vitro (23). Phase I trials in both the
`United Kingdom and the US confirm that, at a dose of
`1000 mg/mz, the reversible hepatotoxicity of N-methyl—
`formamide is dose-limiting and myelosuppression is
`completely absent.2-3 This dose has been defined, as the
`MTD, at which phase II trials have just begun. If activ-
`ity is observed in any tumor type, a repeat phase II
`study in one or more susceptible tumors will be per-
`formed at a level
`immediately below the MTD. This
`procedure will define whether the attainment of toxic-
`ity is necessary for activity.
`
`Phase II
`
`In a phase II trial, the main goal is to assess the ac-
`tivity of the drug in a variety of disseminated malig-
`nancies and to further define the patterns of acute
`toxicity in patients who are homogeneous in diagnosis
`and in better general medical condition than patients in
`phase I. Since large numbers of patients are treated
`during phase II, rarer acute toxic effects often surface
`for the first
`time (24). Also, since cumulative drug
`doses may be appreciable in responding or stable pa-
`tients, phase II provides an appropriate setting for ini-
`tial assessment of chronic toxic effects.
`Several vexing problems are inherent in this process.
`In the first place, since patient numbers and resources
`are finite, it is impossible to explore the activity of
`each drug in each tumor type. A method must be found
`to focus the effort of drug development in a way that
`will minimize the chance of overlooking active agents.
`Accordingly,
`the NCI decided to evaluate all experi-
`
`chVie JG, ten Bokkel Huinink WW, Simonetti G, et al. Phase I trial of
`N-methylformamide (NSC 3051) (NMF). Manuscript submitted to Cancer
`Trea tmcnt Reports.
`3Minutes of the Phase I Working Group Meeting, NCI, Bethesda, MD,
`July 1983.
`
`mental drugs in selected types of cancer. The NCI
`Human Tumor Panel was created in 1975 and included
`lung, colon, and breast carcinomas, and lymphoma, leu.
`kemia, and melanoma. The original intention was to
`match tumors in the human panel with those in the
`preclinical panel, thereby providing information for the
`validation of the preclinical screening program. In addi-
`tion, these classes of human cancer represent the two
`extremes of chemotherapy sensitivity and might be ex-
`pected to exhibit both high sensitivity and high selec-
`tivity. Finally,
`the inclusion of
`the most common
`causes of cancer deaths (breast, colon, and lung can-
`cers) permits the study of large numbers of patients
`and assures that results will have immediate implica-
`tions for the treatment of prevalent cancers. Needless
`to say, evaluation of individual drugs is also carried out
`in tumors other than those in the panel, particularly if
`there is a specific reason to do so. For example, dia-
`ziquone was chemically designed to cross the blood-
`brain barrier and therefore has been extensively eval-
`uated in brain tumors with encouraging results.
`How has activity in the prelinical panel correlated
`with clinical activity? Thus far, we have analyzed the
`results with 13 experimental drugs for which clinical
`and experimental data are available. The correlation of
`activity in each model tumor system with activity in
`the corresponding human cancer is shown in figure 1.
`Prediction of true-negative results (resistance) seems
`fairly reliable across most of the rodent and xenograft
`systems. On the other hand, the probability of predicting
`true-positive results
`(sensitivity)
`is very low. Be-
`cause of the small number of active drugs in humans
`for which complete data are available, no definitive
`conclusions can be drawn. However, even if the pre-
`clinical panel should not turn out to be an accurate pre-
`dictor of response in individual tumor types, overall ac-
`tivity in prelinical screening may still serve as a gen-
`eral predictor of activity in at least one human cancer.
`The aggregated data are, in fact, consistent with this
`notion. This has obviously been the general premise on
`which antitumor screening programs have operated for
`years. Its validity has been widely assumed but has not
`been subjected to direct
`test,
`since drugs are not
`brought to the clinic if screening data are not positive.
`An assessment of the validity of the assumption will be
`afforded by the use of the human tumor stem cell
`assay as a screening tool. The plans are to bring
`selected compounds which are positive in the human
`tumor stem cell assay to clinical trial, even if they are
`negative in the P388 prescreen (26). Obviously, more
`data are needed to determine the ultimate usefulness of
`the panel.
`How has the human panel fared as a predictor of
`clinical efficacy in human tumors other than those of
`the panel? Since 1971, 62 cytotoxic agents have been
`introduced into clinical trials under the sponsorship of
`
`Vol. 68. No. 1,January 1984
`
`This material was {spied
`
`79
`
`

`

`CX-lsc
`Colon xenografts
`
`I+ I-I
`c —I_ ‘ ‘I—T
`o + |
`0
`|
`o
`|
`I
`I
`I
`|
`o "I" _ "I_'I_
`n -
`I
`0
`I
`8
`I
`I
`I
`I
`—T—’_1—'T'
`
`Colon 38
`
`_L:J -I
`I
`|
`I
`c
`0
`0
`o + i
`I
`I
`I __ _____.
`o
`I
`I“_I"
`5
`n —
`I
`i
`3
`i
`‘1__I““—I_
`
`Anguidine, Pyrazofurin, Maytansine, Chlorozotocin,
`Deazauridine, Rubidazone, PALA, Amsacrine
`
`LX—lsc
`Iung xenografts
`
`|
`|
`|
`|
`1_|
`I 0|
`0
`u+l
`n_I_*_l
`|
`g
`I
`I
`|
`'IZI6 I
`~1—_I—I'_
`
`3LL
`Lewis lung carcinoma
`
`|
`|
`|
`"I
`l
`I
`l 0|
`0
`u+l
`n_|___I
`I
`g
`|
`|
`|
`‘IZISI
`T—l—I—
`
`Bis
`melanoma
`
`L1210
`leukemia
`
`—
`0
`
`I
`|
`+
`I
`m
`I
`e_|__~|
`I
`1+|
`0
`|
`I
`a
`I
`|
`nfllfl__I—T_
`o—l
`l
`I
`6
`2
`m
`|
`|
`|
`a
`
`-
`1
`
`I
`l_I__:_|
`|
`e
`I
`I
`I
`u+I
`4
`I
`|
`k__I_____I
`e
`I
`l—I—
`m-I
`3
`I
`I
`1
`1'
`I
`I
`I
`a
`I
`|
`I
`
`Anguidine, Pyrazofurin,
`Maytansine, ChIorozotocin,
`Rubidazone, PALA,
`Amsacrine, DON
`
`Thymidine, Aciacinomycin,
`Anguidine, Chlorozotocin,
`Deazauridine, Rubidazone,
`Indicine N-Oxide,
`Amsacrine, Pyrézofurin
`
`MX-lsc
`breast xenografts
`
`_
`CDSFl
`mammary carc1noma
`
`1
`
`I+| —|
`b ‘1‘ " ‘I
`I
`r + I
`0
`I
`I
`e
`I
`I
`I
`a ‘I‘ “ "I
`|
`s -
`I
`1
`|
`I
`t
`I
`I
`_I—_I—_'I_
`
`6
`
`_L_J -I
`|
`I
`I
`b
`1
`r + |
`|
`I
`e_L_J
`I
`a
`|
`|
`I
`s -
`|
`|
`I
`t
`I
`|
`I
`_I—”_T‘fi_
`
`0
`
`3
`
`5
`
`Anguidine, Pyrazofurin, Maytansine, Chlorozotocin,
`PALA, Amsacrine, DON
`
`Anguidine, Pyrazofurin, Maytansine, Bruceantin, PALA,
`Amsacrine, Mitoxantrone, ChIorozotocin
`
`FIGURE 1.—Correlation of activity of 10 antitumor agents in murine model tumor systems with activity in human cancer. Activity in murine tumors was
`judged according to NCI Decision Network 2 criteria (Goldin A, et al. Eur J Cancer 17:129-142, 1981). Activity in human tumors is defined as a 20% re-
`sponse rate in at least 1 clinical trial with 2 14 evaluable patients.
`
`NCI. Results of an interim analysis of phase II results
`are available for 13 drugs, which were studied in 180
`protocols (table 2). Although these data represent only
`a fraction of the large NCI experience, certain trends
`are apparent. First, significant activity of 2 20% was
`seen. only in the lymphomas,
`leukemia, and breast
`carcmoma (50%, 29%, and 14% of the studies, respec-
`tlvely); most of the results were in the 20%-30% range.
`No drug showed > 20% activity in colon carcinoma and
`melanoma, and only 6% of the lung cancer trials showed
`pOSItive activity. Even with only those drugs which have
`shown activity in at least one tumor type, the overall re-
`sponse rate in colon and lung cancers and melanomas is
`Stlll consistently< 10%.
`
`It is well known that colon cancer and melanoma are
`highly resistant diseases, and that these diseases which
`are consistently refractory to all therapies are of no
`value in screening. Although more data are needed, the
`results to date suggest that inclusion of colon cancer
`and melanoma in the panel may not be useful for
`screening. However, it would seem reasonable to con-
`tinue testing new agents for activity in those common
`and refractory neoplasms until truly reliable screens
`for activity have been defined. Such screens may
`emerge from further analysis of data for clinical trials
`or from advances in the use of in vitro or in vivo lab-
`oratory methods.
`Second, even in intrinsically sensitive diseases like
`
`TABLE 2.-—Outcome of phase II studies in human cancer
`
`M
`
`Disease
`"Breast cancer
`Colon cancer
`Leukemia
`Lung cancer
`Lymphoma
`Melanoma
`
`Total No
`of studies
`30
`38
`21
`47
`18
`26
`
`Response rate
`< 20%
`13 (43%)
`13 (34%)
`9 (48%)
`1 7 (36%)
`5 (28%)
`10 (38%)
`
`13 (43%)
`25 (66%)
`6 (29%)
`27 (57%)
`4 (22%)
`16 (62%)
`
`2 20%
`4 (14%)
`0
`6 (29%)
`3 (6%)
`9 (50%)
`0
`
`———————.—“__—_
`Total
`180
`91 (51%)
`67 (37%)
`22 (12%)
`
`80
`
`TI'I is material was ended
`
`Cancer Treatment Reports
`
`

`

`breast or small cell lung cancer, the number of drugs
`showing activity turned out to be very small. As seen
`in table 3, of the 11 drugs considered in breast cancer,
`only bisantrene showed an overall response rate of
`> 20%. As has been true throughout
`the history of
`medical oncology (27),
`the estimates of activity vary
`widely from trial to trial. For example, with mitoxan-
`trone, response rates ranged from 5% to 28%. This ob-
`servation suggests the well-known importance of fac-
`tors other than drug dose and schedule as major influ-
`ences on estimates of response rate. Again, for mitoxan-
`trone, the deleterious effect of prior therapy on response
`seems to be fairly clear (table 4).
`In fact, the success of the human tumor panel in pre-
`dicting (or ruling out) general patterns of efficacy for
`other human cancers will depend to a large extent on
`what kinds of patients with “panel cancers” are chosen
`for entry in the study. A negative trial of a new drug
`in 20 patients with breast cancer who have failed mul-
`tiple prior regimens tells us nothing about either the
`potential of this drug in a more favorable breast cancer
`population or drug activity in other tumors. Moreover,
`data from earlier eras of cancer chemotherapy cannot
`be used reliably to decide which tumors may be use-
`fully included in a panel without extensive consider-
`ation of how shifting patterns of practice may have al-
`tered important patient characteristics.
`The phase II effort also needs certain administrative
`refinements. Table 5 shows a breakdown by disease of
`patient accrual patterns for negative phase II studies,
`ie, trials yielding a < 10% response rate. Even allowing
`for the histologic heterogeneity of certain primary sites
`such as lung, the extent of overaccrual in some of these
`categories suggests the need for much earlier review of
`the data by investigators and a tighter system of con-
`trol by the statistical offices of cooperative groups. In-
`deed, several groups have already implemented proce-
`
`TABLE 3.—Activity of 11 NCI drugs in patients with breast cancer
`(1975-1980)
`
`
`No. of responding patients/
`Response
`
`Drug
`total evaluable
`rate (%)
`Aclarubicin
`1/48
`2
`Amsacrine
`12/173
`7
`
`Anguidine
`Acivicin
`Bisantrene
`
`Bruceantin
`
`Diaziquone
`Mitoguazone
`Mitoxantrone
`PCNU
`
`1/37
`0/ l5
`18/50
`
`0/15
`
`2/63
`4/104
`16/182
`0/4 5
`
`3
`0
`26
`
`0
`
`3
`4
`9
`0
`
`
`
`3/47Piperazinedione 6
`
`
`
`TABLE 4.—Responses to mitoxantrone in carcinoma of the breast trials
`according to previous treatment
`Response
`No. of previous
`rate (%)
`regimens
`3
`3
`10
`3
`
`Institution ll
`SWOG
`SECSG
`
`5
`
`22
`
`2 1
`
`19
`
`2.6
`
`3
`
`1
`
`1
`
`ECOG
`
`M. D. Anderson Hospital
`and Tumor Institute
`Ohio State
`University
`EORTC
`
`The Royal Marsden
`0
`28
`Hospital
`
`* SWOG = Southwest Oncology Group; SECSG = Southeastern Cancer
`Study Group; ECOG = Eastern Cooperative Oncology Group; and EORTC
`= European Organization for Research on Treatment of Cancer.
`
`dures which should minimize the chances that patients
`will be entered in treatments already shown to be inac-
`tive.
`
`Phase III
`
`Once the activity of a compound is established in one
`or more diseases, subsequent development of the drug
`proceeds along two separate lines. One of these lines is
`to establish the role of the drug in the disease for
`which activity was demonstrated. The endpoints of
`such studies, which are designed to compare the drug
`alone or in combination against standard treatment in
`a randomized fashion, are not only relative activity (eg,
`response rate), but also response duration, survival, and
`toxicity; the ultimate goal is to define the specific con-
`tribution of the drug in the treatment of a particular
`cancer. The data from such trials may be used by
`pharmaceutic firms seeking New Drug Application
`(NDA) approval from the Food and Drug Administra-
`tion (FDA) for marketing purposes.
`In this connection,
`the intense interest in chemical
`analogs of existing active agents poses special chal-
`lenges to clinical drug development. Of 31 drugs devel-
`oped by NCI since 1975, eight have been analogs of
`commercially available or experimental drugs. Until
`very recently,
`the development of analogs proceeded
`along essentially the same lines as that of novel struc-
`tures. Formal prospective comparisons of analog versus
`parent were rarely carried out (28). As a result, little
`direct comparative data exist on the relative merits of
`the various bifunctional alkylating agents, nitrosoureas,
`anthracyclines, or epipodophyllotoxins.
`Surely, if parent and analog have borderline activity
`in a certain cancer, such direct comparisons are prob-
`ably not worth undertaking. Moreover, when such com-
`parisons are worth doing, the trials need to be quite
`
`Vol. 68, No. 1,January 1984
`
`81
`
`

`

`.
`.
`TABLE 5.—Patient accrual onto negative phase II studies us1ng 13 compounds
`—______—_____________—_'__—___—_____________
`No. of studies
`Median
`< 25
`25-50
`2 50
`No. of
`Disease
`Total
`patients
`patients
`patients
`patients
`
`Breast cancer
`10
`1 (10%)
`7 (70%)
`2 (20%)
`32
`Colon cancer
`9
`2 (22%)
`G (66%)
`1 (11%)
`81
`Leukemia
`5
`2 (410%)
`2 (40%)
`1 (20%)
`35
`
`t
`
`:15
`7 (4 7%)
`6 (40%)
`2 (13%)
`15
`Lung cancer (non-small cell)
`(31)
`O
`1
`0
`l
`Lymphoma
`
`
`
`
`
`8 2 (25%) 5 (62%) 1 (12%)Melanoma 85
`
`*Aclarubicin, bisantrene, amsacrine, anguidine,
`piperazinedione, and zinostatin.
`
`acivicin, diaziquone, bruceantin, mitoxantrone, DON, mitoguazone,
`
`l’CNU,
`
`large, because multiple endpoints are involved. Also,
`since current analog programs, especially for platinum
`compounds and anthracyclines, are undertaken to ob-
`viate major organ toxic effects, design of comparative
`trials must permit the simultaneous assessment of rela-
`tive toxicity and efficacy. This task may be particularly
`difficult if one is dealing with equivalent degrees of
`efficacy in the presence of decreased toxicity. In such a
`case, the existence of reduced toxicity is medically im-
`portant only if no material
`loss of efficacy has oc-
`curred. Clearly, accrual should be large enough that
`such a conclusion can be drawn with reasonable power.
`
`Another significant problem with orderly analog de-
`velopment is the current embarrassment of riches: ana-
`logs are much easier to synthesize than to test compre-
`hensively in the clinic. Accordingly, we now have a sit
`uation in which the number of worthy platinum com-
`pounds, anthracyclines, and antifols may actually exceed
`our capacity for rigorous and comprehensive comparative
`trials.
`
`An example of the NCI’s current strategy is the de-
`velopment program for two analogs of cisplatin, CHIP
`and carboplatin. The focus of their development is in
`cancers for which the parent compound is active, but
`not curative, such as carcinomas of the head and neck,
`uterine cervix, and bladder. In these disorders, random-
`ized trials to compare the two analogs are initiated im-
`mediately upon completion of phase I; the superior ana-
`log is then to be compared to the parent compound. The
`trial design includes provisions
`for early stopping
`based on absence of efficacy. Nonrandomized phase II
`trials are implemented in diseases which are less sensi-
`tive to cisplatin, such as non-small cell lung or colorec-
`tal cancer. The results of these trials will show whether
`the spectrum of activity with the analogs is extended
`significantly compared to the parent.
`
`It is also useful to consider within phase III the mul-
`titude of exploratory studies that go on with a new
`agent after conclusion of phase II. These studies in-
`clude:
`incorporation of the new drug into combina-
`
`tions, exploration of alternate routes of administration,
`and use of the agent in high doses accompanied by “res-
`cue” procedures. Such studies, particularly the combina-
`tion trials, constitute a large proportion of clinical
`research in cancer. It would certainly be churlish not to
`acknowledge the many advances that have come from
`such studies, including many of the crucial advances in
`medical oncology (2,3,5). Protection of
`the kidneys
`from platinum-induced damage (13) and current efforts
`with intracavitary chemotherapy (29) and intra-arterial
`chemotherapy (80) are other examples of “post-phase
`II” drug development that either have been proved val—
`uable already or appear particularly promising.
`However, it is equally clear that much of the work in
`phase III, particularly studies with drug combinations,
`has been disappointing in concept, design, or execution.
`Perhaps influenced by the success in certain leukemias
`and lymphomas,
`investigators have all
`too often at-
`tempted to hit
`the home run against
`tumors which
`have repeatedly proven refractory to easy solutions.
`The vast clinical literature in most solid tumors bears
`
`witness to the inefficacy of this approach, at least with
`combinations of drugs having individually low complete
`response rates. At the end of such uncontrolled “pilot”
`studies, one is almost always left with efficacy and
`toxicity data having no clear point of reference. The
`true pilot study does exist, of course, and is an inval-
`uable tool for testing the feasibility of an approach.
`Beyond the question of feasibility, however, a clinical
`trial is always comparative in intent, simply because
`there is always another way to treat the patient. There-
`fore, the issue of a suitable control group will never go
`away, no matter how difficult or inconvenient it is to
`deal with. Whether the control group should always (ie,
`wherever possible) be selected by a random process or
`Whether under certain circumstances nonrandomized
`controls can serve nearly as well is, in a sense, less im-
`portant than the somewhat more basic notion that the
`use of an explicit control group must be much more
`pervasive and sophisticated than is now common prac-
`tice.
`
`82
`
`Th is material was copied
`
`Cancer Treatment Reports
`
`

`

`Coordination of the Process
`
`For many years, the NCI has been the single largest
`contributor to all aspects of anticancer drug develop-
`ment. Because of its multifaceted involvement (from
`organic
`synthesis
`and natural product
`screening
`through phase III clinical trials) and the sheer volume
`of its support for the total effort toward drug develop-
`ment, the NCI has functioned, in effect, as the national
`coordinator of drug development
`in the US. In the
`past,
`the pharmaceutic industry has been less heavily
`involved in the development of antineoplastics than in
`other areas. Recently, however, with the growth of
`clinical oncology as a specialty and the far wider appli-
`cation of chemotherapy in the treatment of cancer, the
`industry has dramatically increased its interest
`in
`cancer.
`
`At the outset, one must recognize that the goals of
`the NCI and pharmaceutic industry share an important
`common feature:
`to define the activity of a new agent
`and make it available to patients as expeditiously as
`possible. It is only in this way that the population at
`large will benefit maximaHy from the fruits of
`research. However, one must also recognize the consid-
`erable differences
`in emphasis
`that
`the NCI and
`pharmaceutic industry often bring to this task. In its
`capacity as a financially disinterested supporter of
`basic and clinical research,
`the NCI wishes to see all
`reasonable steps taken to ensure that the therapeutic
`potential of a new agent is explored fully. By contrast,
`a pharmaceutic firm may be somewhat less inclined to
`support exploratory trials that have no direct role in
`securing an NDA for a new drug, and will strive to get
`a drug to market by the most direct and least costly
`route possible. Once a drug has been approved by the
`FDA,
`the company can deal with the question of
`whether to expand the indications in the postmarketing
`period or, as has been more often the case with anti-
`neoplastic agents, whether to let the evolution of subse-
`quent results work its own effects on the market.
`Obviously, the multiplicity of drug sponsors creates
`both opportunities and problems. The opportunities are
`clear enough: at a time when federal funds for re-
`search are increasingly difficult to obtain, support from
`private sources is sorely needed.
`In addition, many
`pharmaceutic firms now have, in both basic and clinical
`areas, excellent professional staff with the competence
`to formulate and supervise comprehensive drug devel-
`opment programs in cancer.
`The problems deal chiefly with the optimal utilization
`of scarce resources. In recent years, the pharmaceutic
`industry has relied heavily on the same pool of clinical
`investigators who are funded by the NCI via its peer-
`review mechanism, which obviously provides private
`sponsors with an important assurance of competence.
`On the other hand, industry-funded trials may possibly
`
`lead to difficulties in the peer-review process itself, for
`both scientific and financial reasons. At
`least some
`
`trials which are “NDA directed” may not appear very
`exciting or innovative when examined for the renewal
`of an institutional or cooperative group grant. The
`availability of financial support from industry presents
`investigators with the powerful incentives to perform
`trials which may be inherently uninteresting and un-
`likely to move the field forward. The existence of mul—
`tiple sources of private support in addition to federal
`funding creates the neceSSity for very careful account-
`ing of how funds are spent, to avoid the comingling of
`private and federal funds.
`The NCI has attempted to evolve constructive rela-
`tionships with the many pharmaceutic firms involved
`in clinical cancer research. Specifically,
`this involves
`much advanced joint planning to assure the orderly
`development of new agents of mutual
`interest
`in a
`manner which satisfies
`the needs of pharmaceutic
`industry for filing of an NDA with the FDA, while
`simultaneously preserving the autonomy of the clinical
`cooperative groups. As the chief source of funding for
`the cooperative groups

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket