throbber
A History of Cancer Chemotherapy
`(cid:160)
`Vincent T. DeVita, Jr. and Edward Chu
`Cancer Res(cid:160)(cid:160)
`
`2008;68:8643-8653.
`
`(cid:160)
`(cid:160)
`(cid:160)
`(cid:160)
`
`(cid:160)
`(cid:160)
`
`Updated version
`(cid:160)
`
`Access the most recent version of this article at:
` http://cancerres.aacrjournals.org/content/68/21/8643
`
`(cid:160)
`
`Cited Articles
`(cid:160)
`Citing articles
`(cid:160)
`
`This article cites by 88 articles, 19 of which you can access for free at:
` http://cancerres.aacrjournals.org/content/68/21/8643.full.html#ref-list-1
`
`(cid:160)
`This article has been cited by 16 HighWire-hosted articles. Access the articles at:
`
`http://cancerres.aacrjournals.org/content/68/21/8643.full.html#related-urls
`(cid:160)
`
`E-mail alerts
`(cid:160)
`Reprints and
`Subscriptions
`(cid:160)
`Permissions
`(cid:160)
`
`Sign up to receive free email-alerts
`
` related to this article or journal.
`
`To order reprints of this article or to subscribe to the journal, contact the AACR Publications
`.
`pubs@aacr.org
`Department at
`(cid:160)
`To request permission to re-use all or part of this article, contact the AACR Publications
`.
`permissions@aacr.org
`Department at
`(cid:160)
`
`
`
`
`
`Downloaded from Downloaded from cancerres.aacrjournals.org on November 18, 2014. © 2008 American Association for Cancercancerres.aacrjournals.org on November 18, 2014. © 2008 American Association for Cancer
`
`
`
`
`
`Research. Research.
`
`IMMUNOGEN 2108, pg. 1
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`
`A History of Cancer Chemotherapy
`
`Vincent T. DeVita, Jr. and Edward Chu
`
`Yale Cancer Center, Yale University School of Medicine, New Haven Connecticut
`
`Abstract
`The use of chemotherapy to treat cancer began at the start of
`the 20th century with attempts to narrow the universe of
`chemicals that might affect the disease by developing methods
`to screen chemicals using transplantable tumors in rodents.
`It was, however, four World War II–related programs, and the
`effects of drugs that evolved from them, that provided the
`impetus to establish in 1955 the national drug development
`effort known as the Cancer Chemotherapy National Service
`Center. The ability of combination chemotherapy to cure acute
`childhood leukemia and advanced Hodgkin’s disease in the
`1960s and early 1970s overcame the prevailing pessimism about
`the ability of drugs to cure advanced cancers, facilitated the
`study of adjuvant chemotherapy, and helped foster the national
`cancer program. Today, chemotherapy has changed as impor-
`tant molecular abnormalities are being used to screen for
`potential new drugs as well as for targeted treatments. [Cancer
`Res 2008;68(21):8643–53]
`
`Introduction
`In the early 1900s, the famous German chemist Paul Ehrlich set
`about developing drugs to treat infectious diseases. He was the one
`who coined the term ‘‘chemotherapy’’ and defined it as the use of
`chemicals to treat disease. He was also the first person to document
`the effectiveness of animal models to screen a series of chemicals
`for their potential activity against diseases, an accomplishment that
`had major ramifications for cancer drug development. In 1908, his
`use of the rabbit model for syphilis led to the development of
`arsenicals to treat this disease. Ehrlich was also interested in drugs
`to treat cancer,
`including aniline dyes and the first primitive
`alkylating agents, but apparently was not optimistic about the
`chance for success. The laboratory where this work was done had a
`sign over the door that read, ‘‘Give up all hope oh ye who enter.’’
`Surgery and radiotherapy dominated the field of cancer therapy
`into the 1960s until it became clear that cure rates after ever more
`radical local treatments had plateaued at about 33% due to the
`presence of heretofore-unappreciated micrometastases and new
`data showed that combination chemotherapy could cure patients
`with various advanced cancers. The latter observation opened up
`the opportunity to apply drugs in conjunction with surgery and/or
`radiation treatments to deal with the issue of micrometastases,
`initially in breast cancer patients, and the field of adjuvant
`chemotherapy was born. Combined modality treatment,
`the
`tailoring of each of the three modalities so their antitumor effect
`could be maximized with minimal toxicity to normal tissues, then
`became standard clinical practice (1–4).
`
`Requests for reprints: Vincent T. DeVita, Jr. or Edward Chu, Yale Cancer Center,
`Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520. Phone:
`203-787-1010; Fax: 203-785-2875; E-mail: vincent.devita@yale.edu or chueyale@
`yahoo.com.
`I2008 American Association for Cancer Research.
`doi:10.1158/0008-5472.CAN-07-6611
`
`AACR Centennial Series
`
`The Early Period of Cancer Drug Development
`A selected history and timeline of events related to the
`development of cancer chemotherapy is shown in Fig. 1. The first
`four decades of the 20th century were primarily devoted to model
`development. The major limitations of drug discovery were two-
`fold: first, the development of models that could effectively be used
`to reduce the vast repertoire of chemicals to those few that might
`have activity against cancer in humans, and second, the access to
`clinical facilities to test such agents.
`A major breakthrough in model development occurred in the early
`1910s when George Clowes of Roswell Park Memorial Institute (RPMI)
`in Buffalo, New York, Roswell Park Memorial Institute developed the
`first transplantable tumor systems in rodents. This advance allowed
`the standardization of model systems and the testing of larger
`numbers of chemicals. Significant efforts were subsequently focused
`on identifying the ideal model system for cancer drug testing, which
`then became a major thrust of research for the next several decades
`(5–11). The early model systems that were developed included
`Sarcoma 37 (S37), Sarcoma 180 (S180), Walker 256, and Ehrlich’s
`ascites tumor, all carcinogen-induced tumors in mice.
`It was Murray Shear, at the Office of Cancer Investigations of the
`USPHS, a program that was later combined in 1937 with the NIH
`Laboratory of Pharmacology to become the National Cancer Institute
`(NCI), who in 1935 set up the most organized program that would
`became a model for cancer drug screening (7). Shear’s program was
`the first to test a broad array of compounds, including natural
`products, and had both interinstitutional and international collab-
`orations. He ultimately screened over 3,000 compounds using the
`murine S37 as his model system. However, because only two drugs
`ever made it to clinical trials and were eventually dropped because of
`unacceptable toxicity, the program was dissolved in 1953 just as
`discussions began about establishing an organized national effort in
`drug screening. This failure was in part due to the antipathy toward
`the testing of drugs to treat cancer but also to a lack of information
`and experience on how to test potentially toxic chemicals in humans.
`The most excitement
`in this era was generated by the
`introduction of hormonal therapy when, in 1939, Charles Huggins,
`based on an early observation on the effect of estrogens on breast
`cancer made by Beatson in 1896 (12), treated men with prostate
`cancer with hormones and was able to show responses by decreases
`in acid phosphatase levels (13). Although this exciting piece of work
`was an important addition to the systemic treatment of cancer and
`earned Huggins a Nobel Prize, it was not considered to be related to
`the issue of whether chemicals could ever control cancer.
`
`World War II and the Immediate Post-War Period
`Although gases were not used on the battlefield in World War II
`(WWII), a great deal of research was done on vesicant war gases
`(5, 8). The experience in WWI and the effects of an accidental spill
`of sulfur mustards on troops from a bombed ship in Bari Harbor,
`Italy,
`in WWII (14, 15) led to the observation that both bone
`marrow and lymph nodes were markedly depleted in those men
`exposed to the mustard gas. Consequently, Milton Winternitz at
`
`www.aacrjournals.org
`
`8643
`
`Cancer Res 2008; 68: (21). November 1, 2008
`
`Downloaded from
`
`on November 18, 2014. © 2008 American Association for Cancercancerres.aacrjournals.org
`
`
`Research.
`
`IMMUNOGEN 2108, pg. 2
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`
`Cancer Research
`
`Yale, who had worked on sulfur mustards in WWI, obtained a
`contract to study the chemistry of the mustard compounds from
`the U.S. Office of Scientific Research and Development and asked
`two prominent Yale pharmacologists, Alfred Gilman and Louis
`Goodman, to examine the potential therapeutic effects of these
`chemicals. Goodman and Gilman carried out experiments in mice
`bearing a transplanted lymphoid tumor with one compound,
`nitrogen mustard. When they observed marked regressions, they
`convinced their colleague Gustaf Lindskog, a thoracic surgeon, to
`administer nitrogen mustard to a patient with non–Hodgkin’s
`lymphoma and severe airway obstruction. Marked regression was
`observed in this and other lymphoma patients. The initial study
`was done in 1943 but because of the secrecy associated with the
`war gas program, the results were not published until 1946 (16–18).
`The 1943 results set off a burst of support for the synthesis and
`testing of several related alkylating compounds,
`including oral
`derivatives such as chlorambucil and ultimately cyclophosphamide.
`The use of nitrogen mustard for lymphomas spread rapidly
`throughout the United States after the publication of the Lindskog
`article in 1946. If one reads the literature of the time, there was a real
`sense of excitement that perhaps drugs could cure patients with
`cancer (19). Unfortunately, remissions turned out to be brief and
`incomplete, and this realization then created an air of pessimism
`that pervaded the subsequent literature of the 1950s. A cadre of
`academic physicians,
`led by the famous hematologist William
`Dameshek, who having seen apparent success turn to failure could
`never again be persuaded that cancer was curable by drugs (20),
`became harsh critics of a national drug development program and
`the effort to prove that drugs could cure advanced cancers.
`Nutritional research before and during WWII had identified a
`factor present in green leafy vegetables that was important for bone
`marrow function. This factor turned out to be folic acid, which was
`first synthesized in 1937. It was later shown that folate deficiency
`could produce a bone marrow picture reminiscent of the effects of
`nitrogen mustard. Farber, Heinle, and Welch tested folic acid in
`leukemia and they came to the conclusion that it actually acce-
`
`lerated leukemia cell growth (21). Although this observation was
`later proved to be spurious, Farber collaborated with Harriet Kilte
`of Lederle Laboratories to develop a series of folic acid analogues,
`which were in fact folate antagonists, and these compounds included
`aminopterin and amethopterin, now better known as methotrexate.
`Farber subsequently tested these antifolate compounds in children
`with leukemia and, in 1948, showed unquestionable remissions (22).
`Another WWII-related program was the large-scale screening of
`fermentation products by the pharmaceutical industry to isolate
`and produce antibiotics to treat wound infections, based on the
`observations on penicillin. Antitumor effects were examined for
`some agents as well. Penicillin was even initially thought to have
`antitumor properties that were never confirmed. The antibiotic,
`actinomycin D, came from this program. It had significant antitumor
`properties and enjoyed considerable use in pediatric tumors in the
`1950s and 1960s (23). This drug established the initial interest in the
`search for more active antitumor antibiotics, and this effort yielded
`a series of active antitumor antibiotics in common use today.
`Finally, a fourth WWII government effort conducted by the Com-
`mittee on Medical Research of the Office of Scientific Research and
`Development, the antimalarial program, served as an organizational
`model and a source of talent. The success in the search for synthesis
`and production of effective antimalarial compounds in WWII showed
`that a nationally organized, well-supported effort, tightly focused on a
`disease, could yield positive results. Several of the individuals who
`later organized the national effort of the NCI had experience with this
`program in WWII and believed the same kind of effort would yield
`positive results developing drugs against cancer (14).
`The early activity of nitrogen mustard and methotrexate also
`provided a great stimulus for the synthesis of other drugs in
`addition to alkylating agents and antifols. In 1948, the same year
`that Farber showed the antifolate activity of methotrexate in
`childhood leukemia, Hitchings and Elion isolated a substance that
`inhibited adenine metabolism. By 1951, they had developed two
`drugs that would later play an important role in the treatment of
`acute leukemia: 6-thioquanine and 6-mercaptopurine (24, 25).
`
`Cancer Res 2008; 68: (21). November 1, 2008
`
`8644
`
`www.aacrjournals.org
`
`Downloaded from
`
`cancerres.aacrjournals.org
`
`on November 18, 2014. © 2008 American Association for Cancer
`Research.
`
`IMMUNOGEN 2108, pg. 3
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`
`History of Cancer Chemotherapy
`
`activity against a range of solid tumors and, to this day, remains the
`cornerstone for the treatment of colorectal cancer. In retrospect, this
`agent represents the very first example of targeted therapy, which
`has now become the focus of great attention in current cancer drug
`development, although the target in this case was a biochemical
`pathway and not a molecular target. These clinical observations
`increased the interest in chemotherapy and spurred the emergence of
`the R.B. Jackson Laboratories as a major source of inbred mice and
`transplantable tumors, which fostered the establishment of several
`independent screening programs around the world.
`The largest post-war program of drug development before the
`NCI became involved was at the Sloan-Kettering Institute (SKI) in
`New York. Under the leadership of Cornelius ‘‘Dusty’’ Rhoads, nearly
`the entire program and staff of the Chemical Warfare Service,
`including the pioneer clinical investigator David Karnofsky, were
`assembled into the SKI drug development program. The SKI
`investigators used the murine S180 model as their primary screen
`because it was moderately sensitive to known compounds and was
`easily transplanted with nearly 100% success, whereas in Japan,
`Yoshida used an ascites sarcoma model. Additional substantial
`programs were established at the Chester Beatty Research Institute
`in London under Alexander Haddow, the Children’s Cancer
`Research Foundation in Boston under Sydney Farber, and the
`Southern Research Institute in Birmingham, Alabama, under
`Howard Skipper. At that time, the only institutions that had
`facilities devoted to clinical drug testing in cancer patients were the
`Delafield Hospital at Columbia University, Sloan Kettering, the
`Children’s Cancer Research Foundation, and the Chester Beatty (8).
`Rhoads also attracted the interest of the pharmaceutical companies
`by offering to screen and evaluate the pharmacology of submitted
`compounds under special conditions of confidentiality. This
`practice was later adopted into the program of the NCI by Endicott
`as the very important ‘‘Commercial Discreet Agreements,’’ without
`which the industry would not have been willing to cooperate.
`As larger numbers of tumor systems became available, the
`central question for drug screeners at that time was which
`
`Figure 2. Dr. Min Chiu Li. A pioneer chemotherapist who developed new
`curative chemotherapy for metastatic choriocarcinoma and testicular cancer
`(circa 1968).
`
`These thiopurines and other related drugs have been widely used
`not only for acute leukemias but also for other diseases, such as
`gout and herpes viral infections, and as immunosuppressive agents
`in the organ transplant setting. As a result of this seminal work,
`these investigators received the Nobel Prize in Medicine in 1988.
`It was not until the middle 1950s that Charles Heidelberger and
`colleagues at the University of Wisconsin developed a drug that was
`aimed at nonhematologic cancers (26). They identified a unique
`biochemical feature of rat hepatoma metabolism in that there was
`greater uptake and use of uracil relative to normal tissue. Based on
`this observation, Heidelberger ‘‘targeted’’ this biochemical pathway
`by attaching a fluorine atom to the 5-position of the uracil pyrimidine
`base, which resulted in the synthesis of the fluoropyrimidine 5-
`fluorouracil (5-FU). This agent was found to have broad-spectrum
`
`Figure 1 Continued.
`
`www.aacrjournals.org
`
`8645
`
`Cancer Res 2008; 68: (21). November 1, 2008
`
`Downloaded from
`
`cancerres.aacrjournals.org
`
`on November 18, 2014. © 2008 American Association for Cancer
`Research.
`
`IMMUNOGEN 2108, pg. 4
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`
`Cancer Research
`
`Figure 3. Dr. James Holland directed cooperative group studies in childhood
`leukemia (circa 1970).
`
`transplantable tumor was the best at predicting human activity.
`Among those available for use was a murine leukemia induced by
`a carcinogen, Leukemia 1210 (L1210), described by Lloyd Law at
`the NCI (27). This model system was adopted, and its kinetics were
`carefully studied by Skipper and colleagues at Southern Research
`Institute (28, 29) and later by DeVita and colleagues (30). The L1210
`model emerged as the most versatile animal tumor screening
`system and was adopted by the NCI as its primary screen. The
`research that went into the selection of the best screening system is
`reviewed in an article by Goldin and colleagues (5).
`
`The 1950s
`The 1950s were a period of undue pessimism due to the
`disappointment over the failed promise of nitrogen mustard to
`produce durable remissions. This negative view was somewhat
`offset by the discovery of corticosteroids, which were to be used in
`cancer patients but were also quickly found to produce only brief
`responses when used alone (31, 32). Although 5-FU was introduced
`into the clinic in 1958, there were few data of substance about the
`usefulness of this drug until many years later.
`However, the ferment created by the response of acute leukemia
`in children to methotrexate, and the availability of new screening
`systems,
`led to the development of the Cancer Chemotherapy
`National Service Center (CCNSC) in 1955. Although the story of
`how this program was developed is half science and half politics,
`without question it changed the face of cancer drug development
`in the world and changed the NCI and NIH irrevocably. This
`fascinating history is reviewed in detail in the excellent articles by
`Zubrod and colleagues (8) and Goldin and colleagues (5). Given the
`interest in the childhood leukemia data, the National Advisory
`Cancer Council,
`the predecessor to today’s National Cancer
`Advisory Board, convened a panel in 1952 to discuss the subject
`of a national program of cancer drug development and concluded
`that the state of knowledge was inadequate to permit the design of
`a ‘‘crash’’ program. The view that it was premature to develop such
`a program was bolstered by another review in 1954 by a committee
`of the American Cancer Society, chaired by Alfred Gellhorn, a
`prominent academician involved in cancer treatment and Director
`of Columbia’s Frances Delafield Cancer Center (33).
`
`During this time and behind the scenes, the activist and
`philanthropist Mary Lasker,
`in touch with Sydney Farber and
`impressed with the data in childhood leukemia and the antimalarial
`program, had been trying to interest the U.S. Congress in providing
`funds for such a program. In 1954, the Senate Appropriations
`Committee encouraged the NCI to develop a program and provided
`$1 million for cancer drug development. There began a tug of war
`over the proper way to use these funds between members of the
`academic community who preferred that funds be supplied for
`investigator-initiated research and those interested in cancer drug
`screening who preferred a centralized national program. Ultimately
`frustrated by the slow progress, the Senate Appropriations Commit-
`tee, at Mary Lasker’s urging, provided $5 million to NCI with a man-
`date for the establishment of the CCNSC (8). Ken Endicott became its
`first director and was to later become the fifth director of NCI. The
`entire program was set up between May and October of 1955, a
`tribute to Endicott’s organizational skills, and provisions were made
`for commercial discreet agreements with the industry, access to clini-
`cal testing facilities, and the establishment of contracts with organi-
`zations to procure mice and testing sites. In addition, resources were
`made available for pharmacology and toxicology testing and drug
`production and formulation and ultimately an organized decision
`making process called the ‘‘Linear Array with a Decision Network’’
`whereby drugs coursing through the system had to meet specific
`criteria before passing to the next step toward the clinic (34–36).
`As part of the initial development program, the CCNSC set up a
`Cancer Chemotherapy National Committee made up of NCI staff
`with representation from several national organizations as well,
`including the American Cancer Society. This committee then
`established a series of panels to further address each of the major
`issues facing those involved in cancer drug development. This effort
`was the most extensive review of requirements of drug development
`ever conducted. One of the panels of the Cancer Chemotherapy
`National Committee was the clinical panel directed by Gordon
`Zubrod. Out of this effort came the current cooperative group
`program starting with the ‘‘Eastern Solid Tumor Group’’ (now the
`Eastern Cooperative Oncology Group). Subcommittees of this panel
`also addressed the issues of the development of hormone therapy,
`statistical analysis, protocol development, and the design and
`conduct of clinical trials, many of which are still in use today but
`were not in existence in older screening programs like Shear’s at NCI.
`This ensured a wider collaborative effort and provided standardized
`techniques and a stable source of funds, heretofore unavailable, for
`the testing of new approaches to cancer treatment (37, 38).
`The CCNSC programs were supported by contracts, not grants. This
`was the first time contracts had been used at the NCI or NIH for any
`type of program, and it created considerable consternation, which was
`to dog this and a later NCI program, the Special Virus Cancer Program
`(SVCP), for several decades. The use of contracts became synonymous
`with ‘‘targeted research,’’ and was often considered anathema in the
`academic world. Regardless of the quality of the work, it was often
`discounted if it had been supported by contracts.
`In 1966, the CCNSC was incorporated into the NCI structure as
`part of the Chemotherapy Program directed by Zubrod. Now named
`the Developmental Therapeutics Program,
`it was more tightly
`linked to both the extramural clinical trials program and the NCI
`intramural program. This was done over the loud protests of the
`Deputy Director for Science at NIH, Robert Berliner, who feared the
`contamination of the NIH with a contract-supported research effort.
`By 1974, the CCNSC and its successors had grown into an annual
`budget of $68 million and was producing almost 3 million mice
`
`Cancer Res 2008; 68: (21). November 1, 2008
`
`8646
`
`www.aacrjournals.org
`
`Downloaded from
`
`cancerres.aacrjournals.org
`
`on November 18, 2014. © 2008 American Association for Cancer
`Research.
`
`IMMUNOGEN 2108, pg. 5
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`
`bearing transplantable tumors and screening over 40,000 com-
`pounds a year until parts of its effort began to be supplanted by the
`pharmaceutical industry as they began to see an emerging market
`for cancer drugs that worked.
`Still, skepticism surrounded the clinical usefulness of chemo-
`therapy for cancer in the 1950s. A great deal of resources were being
`invested in a controversial effort to develop drugs, yet there was no
`evidence that drugs could cure or, for that matter, even help cancer
`patients in any stage despite some impressive antitumor responses.
`The very rare tumor of the placenta, choriocarcinoma, was the first
`to be cured. The preliminary results of a unique treatment program
`were reported in 1958 (39). The principal architect of the treatment,
`using methotrexate in an unusual way for the time, was Min Chiu Li
`(Fig. 2). The problem was no one was prepared to believe the results
`were significant because the primary site of the tumor was a
`parental hybrid tissue, subject, it was thought, to immunologic
`control. As a sign of the times, after the first two patients went into
`remission, they were presented at NCI Grand Rounds at the Clinical
`Center. The subject of the rounds was ‘‘the spontaneous regression
`of cancer’’ with the speaker being none other than Gordon Zubrod.
`Li was also told that if he persisted in using his radical treatment, he
`would have to forfeit his position at the newly opened clinical
`center. He persisted and was asked to leave (40, 41). Later, when the
`Lasker Prize was given in 1972 to investigators who had participated
`in the studies of the cure of gestational choriocarcinoma, Li shared
`his part of the prize with the person who discharged him. He later
`was to develop the first effective combination chemotherapy prog-
`rams for metastatic testicular cancer (42).
`Clinically, the 1950s ended on the same sour note on which they
`began, but eventually the creation of the CCNSC established one of the
`most successful government programs ever. Although it was often
`criticized (43–46),
`it gave birth to the multibillion-dollar cancer
`pharmaceutical industry. When he was Director of the NCI, Vince
`DeVita was often asked how many drugs came out of the program. The
`answer is, up until 1990, all of them because the CCNSC provided a
`unique central resource, unavailable in medical centers or in industry,
`
`History of Cancer Chemotherapy
`
`Figure 5. Dr. Emil J. Freireich during his days at NCI (circa 1964).
`
`to test, develop, and produce drugs whatever the source. Drugs that
`were not identified in the primary screen itself often were evaluated in
`the ancillary tumor systems, and the necessary toxicology and
`pharmacology for regulatory approval for many drugs was done under
`the auspices of the CCNSC. Clinical studies were then often done
`under contract with the NCI or in one of the national cooperative
`groups. None of this would have been possible in the academic
`medical centers as even today the kinds of resources are not available
`at the majority of university cancer programs nor were these studies
`considered to be worthy of investigator-initiated research.
`
`The 1960s—The Concept of Cure
`In the 1960s, medical oncology did not exist as a clinical
`specialty. Those who were given the task of administering
`chemotherapy at most medical centers were regarded as under-
`achievers at best. The main issue of the day was whether cancer
`drugs caused more harm than good, and talk of curing cancer with
`drugs was not considered compatible with sanity. The prevailing
`attitude toward the use of chemotherapy can only be described as
`hostile. A few vignettes will illustrate this point rather graphically.
`At the medical institution where Vince DeVita began his career,
`the ‘‘chemotherapist’’ was an endocrinologist, Louis K. Alpert, who
`had published one of the early reports on the use of nitrogen
`mustard in lymphomas and administered chemotherapy as a
`sideline. Because of his stern and pointed visage, and because he
`appeared when chemotherapy was to be administered, he was
`referred to by the house staff and the faculty as ‘‘Louis the Hawk
`and his poisons,’’ a designation he took gracefully. Unfortunately,
`poison was the term in general use for anticancer drugs.
`The Francis Delafield Hospital, although connected with
`Columbia University College of Physicians and Surgeons, was
`ultimately denied access to residents and interns from Columbia
`because two successive chairmen of medicine, Robert Loeb and
`Stanley Bradley, did not want their house staff exposed to cancer
`patients receiving these cancer poisons, although their mentor
`would have been the distinguished Alfred Gellhorn. As Alfred
`Gellhorn recently recounted to the authors,1 the otherwise great
`clinician Loeb, a giant in the field at the time, had a blind
`spot when it came to caring for cancer patients and testing
`
`Figure 4. Dr. Emil Frei (circa 1965).
`
`1 Interview with Dr. Alfred Gellhorn (November 26, 2007).
`
`www.aacrjournals.org
`
`8647
`
`Cancer Res 2008; 68: (21). November 1, 2008
`
`Downloaded from
`
`cancerres.aacrjournals.org
`
`on November 18, 2014. © 2008 American Association for Cancer
`Research.
`
`IMMUNOGEN 2108, pg. 6
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`
`Cancer Research
`
`chemotherapy. He was fond of saying to Gellhorn, rather openly,
`‘‘Alfred, you belong to the lunatic fringe.’’ The Delafield Hospital
`program, the first example of a university-based cancer center, with
`many illustrious graduates,
`including Bernard Weinstein, Elliot
`Osserman, John Ultmann, Jim Holland, Paul Marks, Franco Muggia,
`Helen Ranney, and Jack Davidson, was closed in 1971. The leaders
`at Delafield provided the nidus to create a new cancer center at
`Columbia in 1974, after the cancer act in 1971 provided a mandate
`to create new university-based cancer centers.
`At Yale, the first institution to test chemotherapy in humans in
`the modern era, the chemotherapist Paul Calabresi, a distinguished
`professor and founding father in the field, was forced to leave
`because he was involved in too much early testing of new
`anticancer drugs, an exercise as unpopular with the faculty and
`house staff at Yale as it was at Columbia.
`At the Clinical Center of the NCI, where so many of the early
`breakthroughs with chemotherapy occurred,
`the well-known
`hematologist George Brecher, who read all the bone marrow slides
`of the leukemic patients, routinely referred to the Leukemia Service
`as the ‘‘butcher shop’’ at rounds.
`And these are only the stories that can be told. It took plain old
`courage to be a chemotherapist in the 1960s and certainly the
`courage of the conviction that cancer would eventually succumb to
`drugs. Clearly, proof was necessary, and that proof would come in
`the form of the cure of patients with childhood acute leukemia and
`in adults with advanced Hodgkin’s disease.
`By 1960, the L1210 leukemia system had been established as both
`the primary screen and the model for treating acute leukemia. Work
`on L1210, childhood acute leukemia, and Hodgkin’s disease was
`going on in parallel. At the turn of the decade, complete remissions
`were occurring in about 25% of children with leukemia, but with
`
`single agents, they were brief, measured in months. Several
`institutions were cooperating in protocols with a design that hinted
`at cure, not palliation, as an end point. Such studies were in progress
`at RPMI in Buffalo under Jim Holland (Fig. 3), St. Jude’s in Memphis
`under Don Pinkel, Boston Children’s Cancer Center under Sydney
`Farber, Memorial Sloan-Kettering Hospital under Joe Burchenal,
`and the Clinical Center program at the NCI under Emil (Tom) Frei
`(Fig. 4) and Emil (Jay) Freireich (Fig. 5) (46–49). Gordon Zubrod,
`then director of the National Chemotherapy Program, the organizer
`of this effort, played a major role in linking the work of Howard
`Skipper (Fig. 6) on L1210 at Southern Research Institute with the
`clinical programs at the Clinical Center of the NCI and elsewhere.
`A major breakthrough occurred for both leukemia and Hodgkin’s
`disease with the discovery of the activity of the plant alkaloids from
`Vinca rosea at the Eli Lilly Company (50) and discovery of the activity
`of ibenzmethyzin in Hodgkin’s disease (soon to be renamed pro-
`carbazine) by Brunner and Young (51) and DeVita and colleagues (52).
`Furth and Kahn (53) had shown that a single implanted leukemic
`cell was sufficient to cause the death of an animal. At Southern
`Research, Skipper had suggested that to cure L1210, it was necessary
`to eradicate the last leukemia cell because back extrapolations of
`survival after treatment suggested that one surviving cell was
`sufficient to kill a mouse. He offered the ‘‘Cell Kill’’ hypothesis, which
`stated that a given dose of drug killed a constant fraction of tumor
`cells not a constant number, and therefore success would depend on
`the number of cells present at the beginning of each treatment (54).
`This observation changed the existing approach to dosing in the
`clinic in favor of more aggressive use of chemotherapy. In L1210, the
`schedule of administration of drugs was also proving to be
`important. Finally, combinations of drugs, an anathema in medicine
`at the time, were superior to sin

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