throbber
The Anthracyclines: Will We Ever Find a Better Doxorubicin?
`
`Raymond B. Weiss
`
`The anthracyclines are the class of antitumor drugs
`with the widest spectrum of activity in human cancers,
`and only a few cancers (eg, colon cancer) are unrespon-
`sive to them. The first two anthracyclines were devel-
`oped in the 19605. Doxorubicin (DOX) differs from
`daunorubicin (DNR) only by a single hydroxyl group.
`This fact has spurred researchers worldwide to find
`analogs of DOX that have less acute toxicity, cause
`less cardiomyopathy, can be administered orally,
`and/or have different, or greater, antitumor efficacy.
`Five DOX/DNR analogs are marketed in other coun-
`tries, and one (idarubicin) is available in the United
`States. None of these analogs have stronger antitumor
`efficacy than the original two anthracyclines, but there
`are some differences in toxicity. Methods have been
`fashioned to keep the peak plasma level of DOX muted
`to minimize cardiotoxicity, but the only apparently
`effective method available so far (prolonged drug
`infusion) is cumbersome. The bisoxopiperazine class
`of drugs (especially dexrazoxane) provides protection
`against anthracycline-induced cardiomyopathy and has
`much promise for helping mitigate this major obstacle
`to prolonged use of the anthracyclines. The DOX
`analogs being evaluated in the 19905 have been se-
`lected for their ability to overcome multidrug resis-
`tance in cancer cells. Thirty years after discovery of the
`anticancer activity of the first anthracycline, some
`means of reducing anthracycline toxicity have been
`devised. Current studies are evaluating increased doses
`of epirubicin to improve anthracycline cytotoxicity,
`while limiting cardiotoxicity, but at present DOX still
`reigns in this drug class as the one having the most
`proven cancerocidal effect.
`This is a US government work. There are no restric-
`tions on its use.
`
`HE BIFUNCTIONAL alkylating agents
`were the first drugs proven to have clinical
`anticancer activity. One of them, cyclophospha—
`mide, remains one of the most widely used
`drugs today, even though it was synthesized
`nearly 40 years ago. The other most widely used
`
`
`i From the Departments of Medicine, Walter Reed Army
`Medical Center, Washington, DC; and the Uniformed Services
`University of the Health Sciences, Bethesda, MD.
`Address reprint requests to Raymond B. Weiss, MD, Section
`of Medical Oncology, Walter Reed Army Medical Center,
`Washington, DC 20307.
`The opinions erpressed in this article are solely those of the
`author and are not necessarily those ofany government agency.
`This is a US government work. There are no restrictions on
`its use.
`0093- 7754 /92 / 1906-0007
`
`class of antitumor agents today is the anthracy-
`clines, and few patients with malignancies do
`not receive one of the anthracyclines at some
`point in their clinical course. The history of
`these drugs also goes back to the 19505.
`Farmitalia Research Laboratories of Milan,
`Italy (now part of the Montedison Group con-
`glomerate) began an organized effort in the
`mid—19503 to find anticancer compounds pro-
`duced by soil microbes. In 1958, a Farmitalia
`employee collected a random soil sample from
`the grounds of the Castel del Monte, a 12th-
`century castle that is a local tourist attraction
`near Andria in southeastern Italy. From this soil
`sample was grown a newly recognized species of
`Streptomyces, which produced a bright red pig-
`ment. Di Marco isolated an antibiotic from this
`
`fungus1 that had striking activity against a wide
`spectrum of murine cancers.2 It also had antifun-
`gal and antibacterial properties, but
`it was
`selected for clinical development because of its
`antitumor effects. Di Marco et a1 named this
`
`antibiotic daunomycin, using the name (Daunii)
`of a pro-Roman tribe that once resided in the
`Andria region.
`Also in the early 1960s, Dubost et al at the
`laboratories of Rhéne—Poulenc in suburban Paris
`
`independently isolated a new antibiotic from a
`different species of Streptomyces that also pro-
`duced a red pigment.3 They named their new
`antibiotic rubidomycin, using the French word
`rubis, for ruby. On public presentation of their
`new discoveries, both groups of researchers
`recognized that they had identified the same
`substance. The term daunorubicin (DNR) was
`later coined and adopted as the international
`nonproprietary name (INN) to give equal credit
`to both discoverers. By accepted convention, all
`new anthracyclines are given the suffix of
`“rubicin” in the INN system. The name anthra-
`cycline was created by Brockman in the late ‘
`1950s based on the presence of an anthraqui-
`none chromophore and the polycyclic ring sys—
`tem in the chemical structure, which is similar
`to that of tetracycline.
`Clinical trials of DNR began in 1964 in the
`respective countries of origin and at the Memo-
`
`Genentech 2102
`
`570
`
`Hospira v. Genentech
`Seminars in Once/ogy, Vol 19, No 6 (December), 1992: pp 670-686 IPR2017'00737
`
`Genentech 2102
`Hospira v. Genentech
`IPR2017-00737
`
`1
`
`

`

`THE ANTHRACYCLINES
`
`671
`
`rial Sloan-Kettering Cancer Center in the United
`States. At this time, hematologists treating leu-
`kemia and lymphoma were usually the only
`physicians who used drugs for cancer, and DNR
`was tested mostly in patients with these dis—
`eases. This circumstance was actually serendipi-
`tous because DNR was found by Tan to have
`high activity for acute leukemia,4 and this can-
`cer is still the only one for which it is effective.
`By 1967, French and American investigators
`recognized that DNR could produce fatal car—
`diac toxicity,“ which is still the major obstacle
`for prolonged use of anthracyclines.
`Comparison of the structure of DNR to an
`anthracycline predecessor of DNR (rhodomy-
`cin) showed the Farmitalia investigators that
`minor changes in the chemical structure could
`alter the biological activity of this drug class. A
`colleague and collaborator of Di Marco, Ar—
`camone, then began an effort to develop ana-
`logs of DNR that might also have antitumor
`effects. Arcamone et a1 subjected the Streptomy-
`ces that produced DNR to the mutagenic effects
`of N-nitroso-N-methyl urethane and derived a
`strain that produced a different red-colored
`antibiotic.6 Arcamone named this substance
`
`Adriamycin after the Adriatic Sea, which is only
`a few kilometers from the Castel del Monte.
`
`Because Adriamycin was a registered trade
`name, the INN doxorubicin (DOX) was later
`coined for this new agent. Di Marco et al
`showed that DOX had greater activity than
`DNR against some murine cancers and a better
`therapeutic index.7
`DiMarco, who had moved to the Istituto
`Nazionale Tumori in Milan, then took a puri-
`fied supply of Adriamycin for study to his
`clinical colleague, Bonadonna. Serendipity again
`played a strong role because the first patient
`treated with this new drug by Bonadonna had a
`metastatic fibrosarcoma.8 After one dose of the
`
`drug this patient’s pulmonary metastases re-
`gressed. Twenty-five years later, DOX is still the
`most active drug available for treatment of
`sarcomas. The subsequent high antitumor activ—
`ity of DOX observed by Bonadonna et al9 was
`confirmed by Tan et al10 and many others in the
`United States. In 1974, only 6 years after the
`first patient received this drug, DOX was ap-
`proved for marketing in the United States.
`
`DOX remains today the antitumor agent with
`the widest spectrum of antitumor activity. The
`world of oncology owes a large debt of gratitude
`to the Italian investigators, Di Marco, Ar—
`camone, and Bonadonna, who led the research
`
`with these two anthracyclines in the 19605.
`There is a difference of only a single hydroxyl
`group between the chemical structures of DNR
`and DOX,
`in otherwise complex molecules.
`Despite the minor difference in structure, there
`is a great difference in clinical antitumor activ-
`ity. DNR has little activity in carcinomas and
`sarcomas,11 whereas DOX is one of the most
`effective drugs for these cancers. However,
`there are a few cancers where DOX is ineffec-
`
`tive (eg, colon cancer, melanoma, chronic leuke-
`mias, and renal cancer). In addition, the cumu—
`lative cardiotoxicity limits the duration of DOX
`use to approximately 9 months at usual doses,
`and most cancers will develop resistance to it.
`The singular success in developing DOX and
`its limitations in clinical use have been the basis
`
`for investigators worldwide trying to develop a
`better DOX. This research has followed three
`
`major pathways. One has been the creation of
`new anthracycline analogs, in hopes of emulat-
`ing the success of Arcamone. This process has
`continued from the 1960s to the present, and
`will into the future. No one has kept count of
`the new anthracycline analogs synthesized over
`the past 25 years, but it probably numbers well
`in excess of 2,000, and more are reported every
`month. Over 400 analogs have been synthesized
`in one group (the AD series) alone (personal
`communication, Mervyn Israel, July 1992), and
`553 had been evaluated in the screening pro—
`gram at the National Cancer Institute (NCI) by
`1991 (personal communication, Edward Acton,
`September 1991). Another method has been to
`administer some agent in conjunction With DOX,
`either to protect against cardiotoxicity or to
`overcome drug resistance by the cancer. Finally,
`DOX has been modified “mechanically” (eg, by
`the use of liposomes) to minimize heart expo-
`sure to the drug while maintaining antitumor
`efficacy. The major question to be addressed in
`this article is, have (or will) any of these meth-
`ods really given us a better DOX. Although
`there have been many failures in the attempts to
`improve on DOX, in the past few years some of
`
`2
`
`

`

`672
`
`the developments allow a qualified “yes” to this
`question.
`‘
`
`DOXORUBICIN (DOX) ANALOGS
`
`The scientific and commercial success of
`
`Arcamone in adding a single hydroxyl group to
`an active drug and turning it into DOX has been
`the driving force for developing other new
`anthracycline analogs. Analogs have been ob-
`tained from fungi isolated from soil samples or
`rationally synthesized based on known structure-
`activity relationships. A variation of the fungal
`isolation method is to subject the parent Strepto—
`myces organism to mutagens so that new com-
`pounds are created from genetic code modifica-
`tions. Most of this work has been done in
`
`Europe and Japan, and the only Clinically suc-
`cessful analogs developed so far have come
`from Italy and Japan. The focus of such analog
`development has been to find an anthracycline
`that
`is less cardiotoxic,
`is more able to be
`absorbed orally, has less acute toxicity, or has
`activity in cancers resistant to DOX. In the past
`23 years, such a search has led to creation of
`several dozen anthracyclines with promise for
`clinical advantages that unfortunately were not
`borne out in clinical studies. Table 1 lists some
`
`of these agents and the reasons why they are no
`longer in clinical trial. There are other anthracy-
`cline analogs that either are marketed or in
`current trials and have promise for advantages
`over DOX. These will be discussed individually.
`
`Table 1. Some Anthracyclines Tested Clinically and Found to
`Have No Advantages Over DOX
`Name Findings in Clinical Studies
`
`
`Esorubicin
`
`Ouelamycin
`
`Carminomycin Antitumor activity appears inferior to DOX.
`Detorubicin
`Synthesis difficult. No advantages over
`DOX.
`Antitumor activity appears inferior to DOX.
`No less cardiotoxic than DOX.
`Marcellomycin Myelosuppression erratic in phase Itrials.
`No phase ii trials performed.
`Phase I trials showed both acute and chronic
`iron overloading toxicity resulting in he-
`machromotosis. No phase II trials per-
`formed.
`Phase I trials showed both cardiotoxicity
`and nephrotoxicity. No phase II trials per-
`formed.
`
`Rodorubicin
`
`NOTE. None of thesetanthracyclines are in current clinical
`use.
`Abbreviation: DOX, doxorubicin.
`
`RAYMOND B. werss
`
`Idarubicin
`
`The only anthracycline marketed in the
`United States besides DNR and DOX is idaru—
`bicin (IDA). This agent was synthesized by
`Arcamone12 in 1976 and is actually a DNR
`analog because its only difference from DNR is
`the deletion of the methoxyl group at the C-4
`position on Ring D. The INN was derived from
`its Italian name, 4-demetossidaunorubicina. An-
`imal tumor studies were conducted to compare
`IDA with its parent, DNR, and they showed
`that IDA had greater antitumor activity at lower
`drug concentrations.” IDA was found to have
`higher affinity for lipids than other anthracy—
`clines, which suggested that good oral absorp-
`tion was possible. Because both DNR and DOX
`are poorly absorbed orally and must be adminis-
`tered intravenously, oral administration of IDA
`might provide a distinct advantage.
`Phase I trials of IDA in both intravenous and
`
`oral formulations showed that an approximately
`3.5 X greater dose of IDA orally was necessary
`to produce an equivalent myelotoxic effect to
`that of the intravenous drug. Phase II trials with
`both formulations indicated that IDA was ac-
`
`tive in acute leukemias (both myeloid and
`lymphatic) and some carcinomas, notably breast
`cancer.13 As might be expected, the response
`rates were highest
`in patients who had not
`previously received an anthracycline.l4
`Although oral IDA has been studied in pa—
`tients with acute leukemia, there is no advan-
`"tage to this administration route in a disease
`where insertion of a central venous access
`
`device is standard procedure as soon as the
`diagnosis is made. However,
`the activity of
`intravenous IDA in acute myeloid leukemia
`(AML) naturally raised the question, is it any
`better than the parent compound? Only prospec—
`tive randomized trials comparing IDA with
`DNR, both in conjunction with cytarabine,
`could answer this question. There have been
`four such major randomized trials comparing
`these drugs vis-a—vis, each in a slightly different
`dose and/or age populationfiilé The results in
`these studies are not consistent. Two of the four
`
`studies showed a statistically significant advan—
`tage for IDA in complete remission rate and
`two did not. In the three studies where the data
`
`were provided, none showed any difference in
`median duration of response. Finally, two of the
`
`3
`
`

`

`THE ANTHRACYCLINES
`
`673
`
`four studies showed no difference in surviv-
`
`al.15’16 What is puzzling about these trials is that
`the outcomes (response rate, response dura-
`tion, and overall survival) for the IDA-treated
`groups are not really superior to those achieved
`in a large cooperative study using DNR plus
`cytarabine for induction therapy.l7 Moreover,
`the results of the DNR-treated patients in the
`randomized trials are inferior to those in this
`
`cooperative study.”17 On the other hand, when
`there is any difference in the results of the
`randomized trials, IDA is always superior, so
`the clinician is left with uncertainty about the
`question supposedly to be solved by these stud-
`ies: does IDA have a greater efficacy than DNR,
`or is it a more expensive “me too” drug? The
`one conclusive point in these comparisons is
`that there is no meaningful toxicity difference
`between the two analogs.
`DNR is an essential component of therapy
`for acute lymphocytic leukemia (ALL), as it is in
`AML. IDA has activity in ALL at a rate equiva-
`lent
`to that of DNR,” but no randomized
`comparisons of the two analogs have been
`performed in this disease.
`Oral IDA appears to have efficacy in breast
`cancer,14 in contrast with DNR, although DNR
`has had only cursory study in this disease. 11 If an
`oral anthracycline is effective as a palliative
`therapy for breast cancer,
`it would have a
`clearcut advantage over DOX. Does oral IDA
`represent the hoped—for “better doxorubicin”?
`Unfortunately, the answer seems to be no.
`A randomized comparison of oral IDA versus
`intravenous DOX in 76 patients has shown a
`statistically significant,
`inferior response rate
`for the oral IDA.18 In patients who had received
`no prior chemotherapy, the response rate was
`60% for DOX versus 29% for IDA; in patients
`previously treated,
`the response figures were
`29% versus 12%, respectively. This low re—
`sponse rate of oral IDA has been observed in
`single-arm studies also”:20
`A major problem with oral IDA is the varia—
`tion in bioavailability. Stewart et 21121 showed
`that the oral bioavailability ranged from a low of
`12% to a high of 49% in a series of nine
`patients. These data indicate that there can be
`up to a fourfold difference in ability to absorb
`the drug, with commensurate variations in toxic—
`ity (especially hematologic) and antitumor effi—
`
`cacy. This oral bioavailability issue could be the
`explanation for the response rates in breast
`cancer being lower than DOX.”20
`There are some advantages to oral IDA that
`could make it attractive for use in selected
`
`patients. IDA has activity in indolent lympho-
`mas,22 which is a disease where a convenient
`oral palliative drug could be useful as treatment
`for patients not responding to alkylator therapy.
`Myelodysplastic syndrome is another indolent
`disease that could be treated with oral IDA.
`
`IDA might be useful as part of outpatient
`treatment for chronic myelogenous leukemia or
`as maintenance therapy for AML. The apparent
`lower degree of alopecia associated with oral
`IDA18'20 is also a worthy attribute. There also
`may be a somewhat smaller risk of cardiotoxic-
`ity, but a dose limit for heightened risk of such
`toxicity has not been established. However,
`before such oral
`therapy becomes recom-
`mended, it would be worthwhile to study the
`bioavailability problem in more depth. There
`may be phenotypic metabolic variations that
`could be determined before treatment, with
`commensurate adjustments in drug dose to
`minimize toxicity and maximiZe antitumor effi—
`cacy. The IDA patent is held by Farmitalia
`Carlo Erba, and Adria Laboratories, the Amer—
`ican subsidiary of Farmitalia, does not have
`plans at present to pursue marketing of oral
`IDA in the United States but is interested in
`trials that might find oral IDA a niche in cancer
`treatment.
`
`Epimbicin. (EPI)
`
`In their ongoing search for anthracycline
`analogs in the 1970s, Arcamone et a123 modified
`the aminosugar moiety of DOX and created an
`epimer of the C—4’ hydroxyl group on the
`aminosugar (Table 2). The positional change in
`this hydroxyl group is the sole difference from
`DOX. It was selected for further development
`because its murine and human xenograft antitu-
`mor activity was equivalent to DOX, but it had
`less cardiotoxicity.24 These features suggested
`an improved therapeutic index over DOX.
`When phase I testing of epirubicin (EPI) was
`begun in Milan,25 the same every 3-week sched-
`ule used with DOX was also used for this
`
`analog. However, the tolerable dose range estab—
`lished was 70 to 90 mg/m2, which is equimyelo-
`
`4
`
`

`

`Table 2. Anthracyclines Marketed in the United States and/or Other Countries
`Chemical Structure
`Name
`Where Marketed
`Disease Indication
`
`Daunorublcin
`
`Worldwide
`
`Acute leukemias
`
`Doxorubicin
`
`Worldwide
`
`A wide cross-sectlon ol
`carcinomas, lymphomas,
`and sarcomas
`
`Worldwide
`(intravenous only)
`
`Acute leukemias
`
` ‘
`
`rm,
`
`.
`
`o
`
`0
`ll
`c—cH,—l‘oH.‘
`gage “
`u
`\
`on
`
`
`
`Worldwide
`(except U.S.)
`
`A wide cross-section of
`carcinomas, lymphomas,
`and sarcomas
`
`
`
`Japan, France
`
`Carcinomas, lymphomas,
`and sarcomas
`
`ldarubicin
`
`Epirubicin
`
`Pirarubicin
`
`Aclarublcin
`
`Zorubicln
`
`
`
`Japan, France
`
`Acute leukemias and
`non-Hodgkin's lymphomas
`
`Acute leukemias
`
`
`
`NOTE. The dotted line in each chemical structure encircles the point where a difference from daunorubicin exists. The carbon atoms
`are numbered and the rings are lettered in each structure configuration.
`
`5
`
`

`

`THE ANTHRACYCLINES
`
`675
`
`toxic to the 60 to 75 mg/m2 dose range of DOX.
`Identical to DOX, nadir blood counts occurred
`
`10 to 12 days after each dose with recovery by 21
`days. Pharmacokinetic studies26 showed that
`EPI is more rapidly and extensively metabolized
`than DOX with its alcohol metabolite (epirubici-
`nol) being formed to a greater degree.
`In
`addition, EPI forms more glucuronides than
`DOX because of the positional change of the
`C-4’ hydroxyl group. This glucuronidation facil—
`itates the excretion process.26 As a result, the
`terminal elimination phase of EPI is shorter
`than DOX by an average of 10 hours,
`thus
`producing a higher plasma clearance. These
`features of EPI pharmacokinetics are important
`clinically because they may explain the toxicity
`advantage that EPI has.
`Phase II trials of EPI conducted in the 19803
`
`have established the fact that EPI has activity in
`the human cancers for which DOX is active and
`is inactive in the same tumors.”28 Because
`
`DOX is rarely used as a single agent, EPI also
`was evaluated in combination regimens, particu-
`larly for breast cancer. In two large prospective,
`randomized trials comparing EPI with DOX as
`part of combination therapy for metastatic breast
`cancer
`(both groups received cyclophospha-
`mide and 5-fluorouracil to make CAF or CEF),
`there was no difference in response rate,
`re—
`sponse duration, or survival.”30 Therefore, it is
`clear that EPI has not proved to have an
`antitumor efficacy advantage over DOX and is
`disappointing in this regard.
`EPI was selected for clinical development
`because it appeared to be less cardiotoxic than
`DOX. Does EPI have any toxicity advantages
`over DOX? In considering this point one must
`keep in mind the drug doses used in any
`randomized comparison of these two anthracy-
`clines. Are the doses equimolar or equimyelo—
`toxic (a 15 to 20 mg/m2 greater dose for EPI)?
`This point is very important because if equimo—
`lar doses are compared, EPI will always show
`lower toxicity, both acute and chronic. Such an
`outcome was evident in the two studies compar-
`ing CEF with CAP.”30 Both used equimolar
`doses of EPI and DOX, and both showed less
`acute toxicity and cardiotoxicity for EPI. When
`the drugs are compared at equimyelotoxic doses,
`there is little difference in noncardiac toxicity.31
`EPI clearly produces cardiomyopathy that
`
`can even be fatal sometimes-‘2’33 The morpholog-
`ical features of EPI-induced cardiomyopathy
`are identical to those induced by DOX.33 The
`total dose range in which the cardiotoxicity risk
`increases precipitously (akin to the 550 mg/m2
`cumulative dose limit of DOX) is 900 to 1,000
`mg/m2.32“34 This total dose is ideally reached
`with approximately 11 doses of EPI adminis-
`tered over some 9 months,
`if 85 mg/m2 are
`administered at an every 3—weeks schedule.34 If
`a lower dose of EPI is used (eg, 50 mg/m2) when
`it is incorporated into a combination regimen
`such as CEF,”3O then it will ideally take 17
`doses administered over some 12 months to
`
`reach the toxic range. When one considers that
`the median duration of response to CAF or
`CEF regimens in breast cancer is 8 to 10
`months,29:30’35 one can see that most patients will
`have tumor progression and require a change in
`therapy before they have a serious cardiotoxic-
`ity risk from EPI.
`CEF with a dose of EPI equimolar to that of
`DOX in CAF also produces less acute nausea
`and vomiting,”30 an important factor in patient
`compliance with therapy and quality of life,
`apparently without compromising therapeutic
`efficacy. Therefore, EPI seems to provide a
`marginal toxicity advantage over DOX.
`EPI is now widely marketed in Canada,
`Japan, Australia, and Europe, but not in the
`United States, and has largely supplanted DOX
`in clinical use. A New Drug Application (NDA)
`was submitted to the Food and Drug Adminis-
`tration (FDA) in 1985 but was not approved,
`probably because the cardiotoxicity advantage
`of EPI was modest and applied to only a
`minority of patients whose cancer still had not
`progressed after approximately 10 months of
`therapy. The emesis induced by chemotherapy
`is very distressing to patients. If a CEF regimen
`has equivalent efficacy to CAF, but induces less
`vomiting, EPI should be available for clinical
`use in the United States, even if its therapeutic
`spectrum is not different from DOX.
`DOX is usually administered in doses of 60 to
`75 mg/m2 when it is used alone and at 45 to 50
`mg/m2 when used in combination. Increasing
`the DOX dose to 90 to 135 mg/m2 results in
`more total, and more complete, responses in
`breast cancer.36 However, the cumulative dose
`limit for cardiotoxicity is reached more quickly
`
`6
`
`

`

`676
`
`RAYMOND B. WEISS
`
`with such doses, and congestive heart failure
`can result.36 Symptomatic acute mucositis is also
`a problem.
`The maximum tolerated dose (MTD) of EPI
`in the phase I trials conducted 13 years ago25
`was 70 to 90 mg/m2. Drug dose escalation has
`become a topic of intense research interest in
`oncology in the 1980s, and attempts have been
`made to increase EPI doses. These efforts are
`
`based on the premise that the moderate acute
`and chronic toxicity advantages of EPI over
`DOX might allow escalation of EPI doses with
`commensurate enhanced therapeutic efficacy at
`more moderate toxicity cost than similar stud-
`ies36 with DOX. Moreover, the concomitant use
`of bone marrow protective agents (such as
`filgrastim) and cardiotoxicity protectors (such
`as dexrazoxane [DXZ]) may allow even further
`dose escalations of EPI.
`Initial studies of EPI dose escalation in-
`
`creased the dose to 120 mg/mz, with which
`mucositis became dose—limiting, whereas myelo—
`suppression was not.37 Subsequent studies have
`increased the dose to 180 mg/mz,
`in which
`myelosuppression did become dose—limiting and
`caused neutropenic fevers in 23% of 27 pa—
`tients.38 Complete or partial tumor responses
`occurred in 85% of the patients. A study by
`Bastholt et al39 comparing four EPI doses in
`prospective randomized fashion for breast can-
`cer has been preliminarily reported. The re-
`sponse rate was improved by increasing the EPI
`dose from 40 to 60 mg/m2 to 90 mg/m2, but not
`when it was increased to 135 mg/mz. In addi—
`tion, some patients had their dose increased
`after achieving no tumor regression at a lower
`EPI dose, and about 25% then had a respOnse.39
`The higher number of responses achieved
`with both DOX and EPI administered at height-
`ened dOSes are encouraging, but they will not
`necessarily achieve better survival in patients
`with stage IV breast cancer. On the other hand,
`they could be advantageous for patients with
`locally advanced disease. Further work with
`EPI dose escalation using the toxicity pro-
`tectants is ongoing.
`
`Pirambicin (PRA)
`
`Umezawa, at the Institute of Microbial Chem-
`istry in Tokyo, spent a long, illustrious career
`
`‘ searching for antibiotics, particularly those with
`antitumor activity. In 1966 he isolated bleomy-
`cin, and in the 1970s he turned his attention to
`
`finding new anthracyclines. One of the anthracy-
`clines he discovered was a tetrahydropyranyl
`derivative of DOX, which was initially labeled
`THP-Adriamycin.40 The INN now is pirarubicin
`(PRA; the pronunciation of the “pyra” segment
`was internationalized with different spelling).
`Development of this analog has occurred prima-
`rily in Japan and France.
`Preclinical tumor efficacy studies of PRA by
`Tsuruo et al41 showed general equivalence or
`superiority to DOX. Cardiotoxicity studies per—
`formed in France42 indicated a lesser degree of
`cardiotoxicity than DOX in experimental ani-
`mals, and thus PRA was brought to clinical trial.
`The dose and schedule for this analog used in
`phase II trials has been 50 to 70 mg/m2, admin—
`istered at 3-week intervals. Granulocytopenia is
`dose~limiting. PRA has undergone study in
`acute leukemia, lymphoma, sarcoma, and breast
`cancer, and the response rates have been equiv-
`alent
`to those achieved from DOX.”45 The
`
`spectrum and degree of its antitumor efficacy
`appear similar to DOX. When used in a combi-
`nation regimen for breast cancer,46 the response
`rate is equivalent to that of CAP.
`Preclinical studies suggested a lesser degree
`of cardiotoxicity with PRA, but clinical studies
`have not been performed to establish this point.
`In particular, no randomized prospective com-
`parisons of PRA with any other anthracycline
`(such as has been done with EPI31’34) have been
`reported. Also, the cumulative dose at which
`the cardiotoxicity risk becomes significant
`is
`poorly characterized.
`The one attribute of PRA that could incite
`
`clinical interest is the lower rate of total alope—
`cia. When PRA is used as a single agent, total
`alopecia is uncommon, and it does not develop
`as a cumulative toxicity.”45 Even when PRA is
`administered in combination with 5 ~fluorouracil
`
`and cyclophosphamide,46 total alopecia oc-
`curred in only about half of the patients. DOX
`is well known to produce total alopecia in nearly
`every patient treated. Most oncologists have
`encountered patients (especially women) who
`are extremely reluctant to accept any chemother—
`apy that might cause total alopecia. PRA could
`
`7
`
`

`

`THE ANTHRACYCLINES
`
`677
`
`be an acceptable substitute for DOX or EPI
`(with equivalent antitumor activity) that would ‘
`enhance quality of life.
`The patent on PRA is owned by Meiji Seika
`Kiasha, Ltd. of Tokyo, Japan. It is marketed in
`Japan and, under licensing agreements, in Eu-
`rope. No American company has developed any
`interest in pursuing the clinical trials necessary
`to establish a toxicity superiority of PRA over
`DOX. The one attractive feature of causing less
`alopecia may not be commercially promising
`enough to invest the necessary effort and re-
`sources for seeking marketing approval in the
`United States.
`
`Zorubicin and Aclarubicin
`
`isolated by
`Zorubicin (rubidazone) was
`Rhone-Poulenc investigators in France in 1969
`and was found to have activity in acute leuke-
`mia.47 This analog is marketed in France (Table
`2). It was studied by American investigators and
`found to have no advantage over DNR or DOX,
`so no further development for marketing in the
`United States has been done.
`
`Aclarubicin (aclacinomycin) is another ana-
`log found by Umezawa in the mid-1970s. It also
`has activity in acute leukemias and is marketed
`(Table 2) for this cancer in France and Japan.
`Studies in the United States did not show any
`advantages over DNR or DOX, so all clinical
`trials have been closed.
`
`nia, which was rapidly reversible.50 DOX causes
`severe stomatitis at high doses, and during dose
`escalation. This toxicity feature of iododoxorubi-
`cin (IODO) favors the combining of the anthra—
`cycline with hematologic growth factors, such as
`filgrastim or sargramostim, thus allowing fur—
`ther dose escalation with perhaps greater antitu—
`mor activity.51 This analog might even be useful
`as part of a preparatory regimen for marrow
`transplant.
`Only a few phase II studies of IODO have
`been published, and they have not been full of
`promise for this agent“)52 In a trial of patients
`with advanced breast cancer receiving a 70
`mg/m2 dose, the response rate was only 10%,
`and there was negligible activity in colon and
`lung cancer.52 Another trial51 using a dose of 80
`mg/m2 gave a 35% response rate in advanced
`breast cancer, but this rate was achieved at the
`expense of a 34% incidence of grade 4 granulo-
`cytopenia. This hematologic toxicity was also
`more common with repeated treatment. DOX
`can produce a similar response rate as a single
`agent in breast cancer but without such severe
`hematologic toxicity.
`At this point, IODO appears not to provide
`any clearcut advantage over DOX, although
`testing in a variety of cancers has not been
`performed yet. The only promising path for
`further development may be as a constituent of
`marrow ablative therapy.
`
`Iododoxombicin (IODO)
`
`AD-32
`
`This anthracycline is an analog of DOX
`synthesized by Arcamone and colleagues in the
`mid—1980s.48 It was selected for clinical develop—
`ment because it had activity against DOX-
`resistant P388 leukemia, had greater activity
`than DOX in some preclinical tumor lines, and
`had more rapid cellular uptake than DOX. It
`also had less cardiotoxicity in preclinical test-
`ing.49 The sole structural difference from DOX
`is the presence of an iodine atom at the C—4’
`position, instead of a hydroxyl group.
`Phase I trials were performed in Europe in
`the late 19805, and some phase II trials have
`now been completed. A clinically promising
`feature of the toxicity profile, established in
`phase I
`testing, was the fact
`that
`the sole
`dose-limiting acute toxicity was granulocytope-
`
`A series of DOX analogs have been synthe—
`sized by Israel et al, first at Dana-Farber Cancer
`Institute, and subsequently at the University of
`Tennessee. One of the early compounds in this
`series, AD-32, was created in 1973 and had
`greater antitumor activity,
`less cardiotoxicity,
`and less toxicity in general than DOX in preclin—
`ical testing.53 A phase I trial of intravenously
`administered AD—32 was then performed,54 but
`drug formulation and solubility problems pre—
`vented further clinical development.
`AD—32 has recently been resurrected as an
`intravesical treatment for bladder cancer be-
`
`cause it appears not to cause local tissue injury
`if extravasated and is poorly absorbed systemi—
`cally when administered in the bladder.55 Phase
`I trials have been performed,55 and phase II
`
`8
`
`

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