throbber
Breast Cancer Research and Treatment 46: 2550.78, £997.
`(9 1997 Klnwer Academic Publishers. Printed in the Netherlands.
`
`Issues in experimental design and endpoint analysis in the study of
`experimental cytotoxic agents in vivo in breast cancer and other models
`
`Robert Clarke
`
`Vincent T. Lombardi Cancer Center, Georgetown University Medical School, Washington DC, USA
`
`Key words: xencgrafts, breast cancer, cell lines, resistance, cytotoxic drugs, synergy
`
`Summary
`
`Considerable effort has been placed into the identification of new antineoplastic agents to treat breast
`cancer and other malignant diseases. The basic approaches, in terms of model selection, endpoints, and
`data analysis, have changed in the previous few decades. This article deals with many of the issues
`associated with designing in vivo studies to investigate the activity of experimental and established
`compounds and their potential interactions. Endpoints for both in situ and excision assays are described,
`including approaches for determining cell kill, tumor growth delay, survival, and other estimates of
`activity. Suggestions for approaches that may limit the number of animals also are included, as are
`possible alternatives for death as an experimental endpoint. Other concerns, such routes for drug
`administration, drug dosage, and preliminary assessments of toxicity also are addressed.
`Statistical
`considerations are only briefly discussed, since these are addressed in detail in the accompanying article
`by Hanfelt (Hanfelt J], Breast Cancer Res Treat 46:279—302, 1997). The approaches suggested within this
`article are presented to draw attention to many of the key issues in experimental design and are not
`intended to exclude other approaches.
`
`Introduction
`
`The disseminated nature of breast cancer and the
`
`tumors are the
`development of crossresistant
`primary causes of failure of current therapies. By
`the time many tumors are detected, there is a high
`probability that metastatic lesions will be present,
`many of which may already contain resistant sub-
`populations [1]. Not surprisingly, there is sub-
`stantial
`interest
`in the identification of novel
`
`cytotoxic and endocrine agents for the treatment
`of breast cancer.
`
`A systemic approach is required to eradicate
`
`the majority of metastatic breast disease, and this
`remains primarily in the form of cytotoxic chemo-
`
`therapy or endocrine manipulation. The latter
`began with the initial studies on oophorectomy by
`Beatson [2] and was followed by the administra—
`tion of high dose estrogens. These were largely
`replaced by the development of antiestrogens,
`
`Address for correspondence and ofiprints: Robert Clarke PhD, Vincent T. Lombardi Cancer Center and Department of Physi—
`ology & Biophysics, W405A The Research Building, Georgetown University Medical School, 3970 Reservoir Road NW,
`Washington DC 20007, USA; Telephone: (202) 687-3755; Fax: (202) 687-7505; email: Clarker@gunet.georgetown.edu
`
`Genentech 2052
`
`Celltrion v. Genentech
`
`|PR2017-01122
`
`Genentech 2052
`Celltrion v. Genentech
`IPR2017-01122
`
`

`

`256
`
`R Clarke
`
`Table 1. Several of the human breast tumor cell lines used
`as xenografts in the current NCI drug screening program*
` Cell Line ER status Cltation
`
`
`
`BT7549
`H5578T
`MCF-7
`
`MCF7/ADR
`(multidrug resistant)
`
`MDA-MBw231
`(metastatic)
`MDAiMB—ABS
`(metastatic)
`
`negative
`negative
`positive
`
`negative
`
`**
`(106)
`(107)
`
`(39,40,106)
`
`negative
`
`(108,109)
`
`negative
`
`(108 110)
`
`
`
`positiveT47D ( l l l)
`
`
`
`* Kindly provided by Dr. Joseph Mayo.
`** There is no primary citation for this cell line listed by
`the provider (ATCC, Roekville, MD), who indicate in their
`“Catalogue of Cell Lines and Ilybridomas“ that the cells
`were derived from a papillary invasive ductal tumor in a
`72-year old patient. Metastatic disease was found in three
`of seven regional lymph nodes. The originators were Dre
`W.G. Coutinho and E.Y. Last'argues. A discussion of other
`human breast cancer cell
`lines can be found elsewhere
`[112].
`
`initially the triphenylethylene Tamoxifen [3] and,
`more recently, introduction of the steroidal anti-
`estrogen ICI 182,780 [4]. Other endocrine strate-
`gies include the use of inhibitors of estrogen
`biosynthesis, primarily aromatase inhibitors, and
`the use of LHRH agonists and antagonists.
`The use of chemotherapeutic agents in the
`management of neoplastic disease began with
`Rhoad’s description of the use of nitrogen mus—
`tard for the treatment of Hodgkin’s lymphoma.
`[5]. The number of cytotoxic agents available has
`increased substantially over the intervening years,
`with breast
`tumors generally exhibiting good
`overall response rates to several cytotoxic drug
`combinations. While chemotherapy can produce
`gains
`in overall survival, most patients with
`metastatic breast cancer will eventually recur.
`Many reasons may account
`for
`this
`failure,
`including poor dose scheduling,
`inappropriate
`combinations of drugs and the emergence of cell
`populations resistant to the antineoplastic agents.
`Animal models provide one approach for the
`
`the
`optimization of drug scheduling and for
`identification of novel compounds that exhibit
`promise in such in vitro prescreens as
`those
`currently utilized by the National Cancer Institute
`[6]. For many years a primary in vivo screen,
`utilizing the L1210 and P388 mun'ne leukemias,
`was an integral part of the NCI‘s preclinical drug
`development program. However, the lack of a
`significant representation of solid human tumors
`drew criticism, and partly explained the weakness
`of the screen to identify new agents active against
`the more common solid tumors, including breast
`cancer.
`Perhaps not surprisingly,
`the screen
`appears to have been more successful in identify
`ing agents active against hematologic malignan—
`cies. Some drugs with well established clinical
`efficacy (busulfan, hexamethylmelamine) fail to
`demonstrate substantial activity in the L1210/P388
`screen [7,8].
`
`The P388 and L1210 in viva screen has largely
`been replaced by panels of disease specific human
`tumor cell
`lines
`(prescreen)
`and xenografts
`(primary screen). While there are relatively few
`ideal in vivo models for breast cancer, several of
`the available human cell lines fulfil some of the
`
`requirements for screening, 6.3., stable phenotype,
`high tumor take rate, predictable and reproducible
`kinetic properties. Several of those currently used
`by NCI are shown in Table 1.
`
`Some considerations for choice of model,
`scheduling, dosage, and endpoint will be dis—
`cussed below. The discussions and issues raised
`
`are to draw attention to different approaches to
`experimental design. AS such, these Should be
`
`considered in the light of the other articles on
`experimental design and data analysis in this issue
`and elsewhere. Some issues are generic, while
`others are more directly applicable to screening
`cytotoxic activities, and may be of lesser rele-
`vance to testing chemopreventive and endocrine
`agents.
`It is hoped that the topics discussed will
`assist
`investigators to consider key issues in
`experimental design. However,
`these are pro-
`vided only as suggestions. There are many ways
`to identify potentially active compounds and drug
`combinations, and these are constantly being
`
`

`

`modified by improvements in both cellular models
`and approaches to the use of animals in biomed-
`ical research.
`
`Log cell kill and tumor kinetics in cytotoxic
`
`cancer chemotherapy
`
`Early attempts to modify cytotoxic therapies were
`
`The initial criteria for de—
`largely empirical.
`signing cytotoxic therapies were based on the
`observations of Skipper, who demonstrated that
`
`cytotoxic drug-induced cell kill follows a similar
`kinetic pattern to that established by Arrhenius at
`the turn of this century for the killing of bacteria.
`The log cell kill hypothesis states that cytotoxic
`drugs kill cells by first order kinetics [9]. Thus.
`a constant proportion of the cells will be killed
`regardless of the size of the cell population. For
`many cytotoxic drugs a 4—log cell kill is achiev—
`able and would eradicate a tumor population of
`103 cells and have a one in ten chance of elimin—
`ating a population of 104 cells [10]. However,
`clinically detectable primary tumors have a cell
`population frequently in excess of 109 cells.
`Many metastases also could contain cell popula—
`tions greater than 104 cells. While theoretically
`sound,
`log cell kill can be affected by several
`biological parameters, including the presence of
`de novo resistant cells,
`the degree of tumor
`vascularity, and other factors that may affect drug
`perfusion and metabolism.
`The principles elucidated by Skipper were
`further modified by Norton & Simon, who ap-
`
`plied Gompertzian growth kinetics to tumor cell
`populations [11]. This clarified the inverse re-
`lationship between growth fraction and tumor
`size. The ability of a cytotoxic drug to inhibit
`tumor cell growth was determined to be directly
`related to the tumor’s growth rate, which also is
`related to tumor volume [12]. The tumor mass
`
`killed by a cytotoxic treatment is proportional to
`the growth fraction multiplied by the total tumor
`volume [12].
`
`A GOmpertzian kinetic growth pattern pro
`duces a growth fraction that ultimately decreases
`
`Breast cancer models for drug screening
`
`257
`
`exponentially with time. This inverse relationship
`between tumor size and growth fraction implies
`that micrometastases should be more kinetically
`sensitive to cytotoxic chemotherapy than larger
`
`tumor masses [13,14]. Thus, early intervention
`when the tumor mass is small should provide the
`
`greatest opportunity for induction of remission.
`Metastases tend to exhibit a more rapid tumor
`doubling time (TD) than primary tumors, particu—
`larly for the common solid tumors like those of
`the breast [15].
`There are a number of factors that contribute
`
`to the apparent TD of any tumor. These include
`the rate of cell production, the size of the growth
`fraction, cell recruitment from G0, and the rate of
`cell loss. Cell loss includes shedding of cells into
`other compartments. e.g.. metastasis. differenti-
`ation to a non—proliferating cell type, or entry into
`
`prolonged G0 and apoptotic cell death. The high
`proportion of nonwproliferating or normal cells
`
`present in many solid tumors, and the frequently
`high rate of cell loss, generally produce tumors
`with a long TD. The rate of cell loss may be as
`high as 80% of the rate of cell production [16].
`
`Growth kinetics in human tumors and animal
`models
`
`The kinetic parameters of tumor growth represent
`one of the major differences between animal
`models and the human disease. While Gompertz~
`ian kinetics apply to experimental
`tumors and
`
`those in patients, the TDs and growth fractions are
`frequently quite different. For example, many
`human breast tumors exhibit long TDs and often
`small relative growth fractions.
`In marked con—
`trast, human breast tumor xenografts generally
`have short TDs and high growth fractions. Estro—
`gen-treated MCF~7 tumors (ER—positive) have TDs
`of approximately 1042 days [17-19] compared
`with over 100 days for many tumors in patients
`
`[20]. We have generated several estrogen-in-
`dependent MCF-7 variants. These have TDs as
`long as 100 days when grown in the absence of
`
`

`

`258
`
`R Clarke
`
`estrogen, but grow as rapidly as parental MCF—7
`tumors in estrogen-supplemented mice [17-19].
`MDA435/LC‘C‘6 cells (ER-negative, ascites variant
`
`of MDA-MB-435) growing as solid tumors have
`much shorter TDs of 3-5 days [21]. Most of the
`breast cancer xcnografts used in the current NCI
`screen have mean TDs of 210 days.
`The differences in kinetic properties between
`xenografts and tumors in patients would tend to
`make the xenografts more sensitive to agents with
`a strong cell cycle/cell phase specificity. For the
`purposes of a primary in viva screen for novel
`agents, a limited overestimate of activity may not
`be a major concern.
`For studies to optimize
`scheduling or combinations of established drugs,
`the relative sensitivity of the in viva screen is a
`concern only if the model is either too sensitive
`or too resistant to a combination, when it will
`become difficult to assess interactions. Most of
`
`these concerns are readily addressed by a careful
`choice of in. viva model(s). Various schedules
`
`have already been identified for the established
`drugs, examples are provided in Table 2.
`
`the dose response relationship and dose
`intensity
`
`The relationship between treatment and response
`is described by:
`
`k—Cxt
`
`res—
`t = time. Thus,
`where C m concentration;
`ponse should be approximately equivalent where
`C x t values (area under the concentration time
`
`curve) are equivalent. This can enable the design
`of clinically relevant in vitro analyses of estab«
`lished drug combinations based on pharmaeokin-
`etic measurements previously obtained in patients
`or animals.
`Clinical studies can utilize the
`
`reasonable across-species dosage relationship of
`mg/m2 to estimate dose from data obtained in
`preclinical animal screening. The doses may re—
`
`quire some further modification, since the serum
`half-life of some drugs can be longer in man than
`in rodents [22]. One approach is to use 1/ 10th
`
`the maximum tolerated dose (MTD) in rodents as
`
`the approximate starting dose for a Phase I trial in
`humans [93]. Where possible, it may he better to
`
`use a dose that produces comparable pharmaco-
`kinetics, since this can increase the predictability
`of the mOuse xenograft to human tumor model
`[24].
`
`The pharmacokinetics for cytotoxic drugs are
`frequently similar in mice and men [25]. For
`many drugs, the mouse LD10 also approximates
`the MTD in humans when expressed as mgl'm2
`[26]. However, there are exceptions. The C x t
`
`values at the LD10 are higher for mitomycin C,
`vincristine, and cyCIOphosphamide, and lower for
`methotrexate and 5-fluorouracil,
`in mice when
`
`compared with humans (reviewed in [26]).
`In clinical practice. a narrow therapeutic index
`is frequently responsible for the reduction of
`dosage due to side effects. However, the steep
`dose response curve for most cytotoxic drugs im—
`
`plies that even a small perturbation in dosage may
`produce a significant change in response. The
`effects of alterations in dosage on clinical res—
`
`It has
`ponse has been widely reviewed [22,27].
`been suggested that a major contributing factor to
`the failure of many treatments is the ad hoc re—
`
`duction of drug dosage [22].
`It has been widely acknowledged that the most
`effective treatments involve a high dose intensity
`chemotherapeutic regimen. This is partly based
`on the steep dose response relationship for most
`cytotoxic
`drugs
`and
`various other
`clinical
`observations. Dodwell er al. [27] have reviewed
`the published data, and reexamined the role of
`dose intensity in reSponse, for a number of the
`more common malignancies. While they con-
`clude that high intensity regimens can produce
`significant advantages in disease free survival,
`clear demonstrations of increased overall survival
`
`are obtained much less frequently. The relation-
`
`ship between dose intensity and response often
`varies with both drug, tumor model, and disease.
`Animal models provide a safe and logical means
`
`to explore this and related issues, rather than
`attempting to identify appropriate or potentially
`dangerous schedules directly in patients.
`
`

`

`Table 2. Examples of dosages and schedules for several established cytotoxic agents. The tumor models against which the
`drugs were tested were of various origins, and not exclusively breast. Some drugs that are not widely used in breast cancer
`are included, since these may be useful as controls for establishing the extent to which a tumor model reflects the human
`disease. Toxicity can vary with strain, sex, age and other parameters, and the information in this Table reflects that diversity.
`
`Breast cancer models for drug screening
`
`259
`
`Drug
`
`Adriumycin
`
`Ara-C6
`
`BCNU
`
`Methyl-CCNU
`
`Cyclophosphamide
`
`S-Fluorouracil
`
`lfosfamide
`
`Melphalan
`
`Methorrexate
`
`Mitomycin C
`
`Ctr-Platinum
`
`Taxol
`
`Taxotere
`
`Vinblastine
`
`Dosel
`
`6 nag/kg
`6 mg/kg
`6.8 mg/kg
`8.5 mg/kg
`24 Inglm2
`
`40 rug/kg
`50 mgfkg
`
`18 rug/kg
`
`18 mgfkg
`
`35 mgfkg
`60 mglkg
`100 rug/kg
`143 rug/kg
`200 mgfkg
`286 rug/kg
`290 mgfkg
`
`32 mglkg
`40 mg/kg
`50 mgfkg
`60 rng/kg
`180 rnglrn2
`
`150 mgfkg
`300 rug/kg
`
`12 mgfkg
`12 mg/kg
`
`4 mg/kg
`15 mg/kg
`20 mg/kg
`
`2 mg/kg
`2.5 rag/kg
`4.5 mg/kg
`5 mg/kg
`18 rngfrn2
`
`4 mgfkg
`4 rug/kg
`7.5 mglkg
`10 mgfkg
`2’? mgi'm2
`
`20 rug/kg
`
`15 mg/kg
`
`3 mg/kg
`
`Route
`
`i.v.
`i.v.
`i.p.
`i.p.
`i.v.
`
`so.
`i.p.
`
`i.p.
`
`i.p.
`
`i.p.
`i.p.
`LV.
`i.p.
`i.p.
`i.p.
`i.p.
`
`i.p.
`i.p.
`i.p.
`i.v.
`i.p.
`
`i.v.
`i.v.
`
`i.p.
`i.p.
`
`i.p.
`i.v.
`i.p.
`
`i.v.
`i.v.
`i.p.
`i.p.
`i.p.
`
`i.p.
`i.v.
`i.p.
`i.p.
`i.p.
`
`i.p.
`
`i.v.
`
`i.v.
`
`Schedule2
`
`4,5,9
`1,5,9
`single dose
`single dose
`single dose
`
`24 hr infusion
`1-7
`
`single dose
`
`single dose
`
`single dose
`0-4, '7—11
`1,5,9
`single dose
`1,15
`single dose
`single dose
`
`5,6,7,8
`1-4, 15-18
`0-4
`1,5,9
`single dose
`
`single dose
`single dose
`
`single
`0,4
`
`1,4,8,11,15,18
`1-5
`03
`
`1.15
`single dose
`single dose
`single dose
`single dose
`
`1,5,]0
`1,5,10
`single dose
`single dose
`single dose
`
`single dose
`
`4,6,8
`
`1,15
`
`Toxicity
`
`N133
`ND
`none4
`LD5
`MTD
`
`none
`alopecia
`
`LD
`
`MTD7
`
`alopecia
`ND
`ND
`none
`LD
`LD
`none/LD
`
`ND
`LD
`ND
`ND
`MTD
`
`none
`none
`
`ND
`ND
`
`LD
`ND
`ND
`
`LD
`LD
`none
`ND
`MTD
`
`ND
`ND
`none
`ND
`MTD
`
`none
`
`MTD
`
`LD
`
`Citation
`
`[67,113]
`[1 14]
`[21]
`[21]
`[25]
`
`[115]
`[1 16]
`
`[21]
`
`[71]
`
`[1 1.6]
`[117]
`[114]
`[118]
`[119]
`[118]
`[81]
`
`[72]
`[I 19]
`[11.7]
`[114]
`[25]
`
`[7'7]
`[77]
`
`[120]
`[120]
`
`[119]
`[119]
`[117‘]
`
`[119]
`[121]
`[21]
`[122]
`[25]
`
`[123]
`[123]
`[21]
`[123]
`[25]
`
`[2]]
`
`[67]
`
`[119]
`
`1 = Dose/injection; 2 = Schedule is given as days unless otherwise indicated; 3 = Not defined or not described; 4 = None reported
`or no deaths; 5 : Lethal dose (one or more deaths attributed to drug~induced toxicity): 6 = 1-l3—D-arabinofuranosylcytosine: 7 =
`Approximate MTD as defined by the investigators.
`
`

`

`260
`
`R Clarke
`
`General considerations in experimental
`design
`
`Choice of host for xenografis
`
`There are several considerations relating to the
`choice of immunocompromised host,
`including
`the degree of immune-competence and the ability
`of the strain to support the growth of the tumor.
`For syngeneic tumors the choice of host should be
`obvious. However, the choice of host for xeno-
`
`grafts is more complex. The host not only should
`facilitate the growth of the tumor but also should
`enable the tumor to maintain a biologically rele-
`vant phenotype.
`It also is useful if the selected
`model enables some estimation of the selectivity
`of drug action and the determination of potentially
`lethal toxicities. For most of the breast cancer
`
`cell lines/xenografts available, the nil/nu mouse is
`sufficient, irrespective of the background strain
`[28].
`
`The potential for immunological modulation
`to contribute to tumor response to combination
`chemotherapy is an important consideration. This
`is particularly relevant when the combination
`includes biological response modifiers that can
`influence the activation of cells associated with
`
`interferons and
`cell mediated immunity, e.g.,
`interleukins.
`In some cases, it may be desirable
`to attempt to either eliminate or exclude effects
`on the immune competence of the host. Thus, the
`choice of an appropriate immune—deficient strain
`may become paramount. Since the different im—
`munobiologies of available rodent hosts have been
`recently reviewed [28], they will not be further
`discussed.
`
`Choice of appropriate tumor model
`
`The model used for the screening of a drug sus-
`pected of activity against a particular
`tumor
`should reflect
`the biological properties of that
`tumor as closely as can reasonably be achieved.
`For example, a drug active against a leukemia
`with a short TD, high growth fraction, and
`
`relatively short cell cycle time would be less
`likely to demonstrate activity in a screen against
`a breast tumor with a longer TD, small growth
`fraction, and long cell cycle time. Thus, choice
`of an appropriate model to screen combinations of
`agents with known pharmacological and kinetic
`requirements
`should (where possible) closely
`reflect
`the major biological properties of the
`human disease. To some extent the model also
`
`should reflect the requirements of the drug. For
`example, there would be limited value assessing
`a drug expected to show no crossresistance to P—
`
`glycoprotein without including a model that ex—
`presses P-glycoprotein.
`Selecting a breast tumor model for screening
`cytotoxic compounds can be problematic. Rela-
`tive to the murine ascites models, most solid
`
`breast tumors exhibit a relatively slow TD. For
`example, MCF-7 tumors have a TD of approxi-
`mately 10-12 days when growing in appropriately
`estrogen supplemented animals [19]. While this
`may be acceptable for screening antiestrogenic or
`chemopreventive agents, cstrogenie supplementa—
`tion can alter the activity of some cytotoxic drugs
`[29,30]. In general, the most rapidly proliferating
`human breast tumor xenografts do not express
`estrogen receptors. MDA435/LCC6 tumors have
`TDs of 2-3 days.
`Since the parental cell
`line
`(MDA—MB-435) was obtained from a patient who
`had not received chemotherapy, these models may
`be useful in screening cytotoxic agents for activity
`against breast cancer [21]. The MDA-MB—231
`cell line also has a comparably rapid TD in vivo
`(R. Clarke, unpublished observations). The more
`rapid TDs of ER-negative xenografts broadly
`reflect
`the characteristics of such tumors in
`
`patients, where ER—negative tumors tend to have
`shorter TDs [31].
`From a purely practical viewpoint, relatively
`slowly proliferating breasr tumors can produce
`significant problems in logistics and experimental
`design. The slower growing tumors frequently
`exhibit significant intertumor variability and can
`require substantial numbers of animals to enable
`meaningful statistical analysis of data.
`Deter—
`mining the period at which a tumor is considered
`
`

`

`"cured" also can become problematic. Even the
`more rapidly proliferating solid tumors with TD 2
`48 hr may require up to four months of post treat-
`ment'observation to establish "cure" [32].
`
`It is unlikely that any one tumor model will
`adequately represent the maior biological charac-
`teristics of a particular malignancy. Thus, the use
`of a series of tumors (where appropriate/available)
`may be. required to determine the sensitivity of a
`particular neoplastic disease to a either a single or
`a combination chemotherapy regimen. However,
`this must be considered in the context of reducing
`
`animal usage, cost, and the value of the additional
`data obtained.
`
`For breast cancer, there are several potential
`models available for screening (Table 1). Most of
`
`the ER—positive models require estrogenic supple—
`mentation for tumorigenicity or maximal growth.
`We have generated ER—positive models that will
`grow without supplementation, but the respective
`TDs are relatively long [19]. While this may be
`more representative of breast tumors in general,
`this characteristic is inappropriate for screening
`cell cycle or cell phase specific cytotoxic com-
`pounds. We also have developed an ascites mod-
`el based on a variant of the MDA—MBABS cell
`
`line (MDA435/LCC6). The pattern of response to
`a variety of cytotoxic drugs appears to reflect
`closely that seen in breast cancer patients [21].
`For example, breast cancers in general respond
`poorly to nitrosoureas [33], as do MDA435/LCC6
`ascites to BCNU. Etoposide also does not pro—
`
`duce long term Survivors, and this drug generally
`has been ineffective as a single agent in breast
`cancer [34,35]. Adriamycin [34], mitomycin C
`[36], and taxo] [37] are among the most effective
`single agents
`in previously untreated breast
`cancer, and all of these drugs produced long term
`survivors in mice bearing the MDA435/LCC6
`ascites.
`The characteristics of several breast
`
`cancer xenografts have been reviewed elsewhere
`[28].
`Another example of the choice of tumor
`model
`is in studies to evaluate P—glycoprotein
`reversing agents, compounds which may have sig-
`nificant potential in some breast cancer patients
`
`Breast cancer models for drug screening
`
`261
`
`[38]. For these types of analyses the choice of
`tumor model
`is critical. Cells to be used as
`
`xenografts should be transfectants rather than
`selected for
`resistance,
`since
`selection can
`
`produce multiple unrelated resistance mechanisms.
`For example, MCF—7ADR (selected for resistance
`to adriamycin [39]), but not MDRlutransdnced
`MCF-7 cells (CL 10.3), are cross resistant to
`Tumor Necrosis Factor [40]. Since both adria—
`
`mycin and Tumor Necrosis Factor can inhibit
`cells by the generation of free radicals [41,42],
`this cross resistance in MCFFJ'ADR
`cells suggests
`
`the presence of functional adriamycin resistance
`mechanisms in addition to P—glycoprotein, includ-
`ing altered expression Uf ilianganous superoxide
`dismutase [40]. These cells also exhibit increased
`
`glutathione transferase and topoisomerase 11
`activities [43,44]. The use of transfected cells
`
`allows for a clearer interpretation of the data.
`Cells concurrently expressing multiple resis~
`tance mechanisms may more closely reflect the
`drug resistance that occurs in patients [38], but
`interpreting responses in a mechanistic light may
`be difficult. This does not invalidate their use
`
`where the purpose is simply to sereen compounds
`for potential antineoplastie activity.
`indeed, the
`choice of a series of models that are too sensitive
`
`will likely identify compounds with limited activ—
`ity in patients, whereas active compounds identi—
`fied in otherwise resistant models may have a
`higher probability of being active in patients [45].
`This is likely to be true in principle, but
`the
`extent
`to which it applies will depend upon
`whether the resistance mechanisms operating in
`the tumor model contribute significantly to the
`
`resistance phenotype in patients.
`It
`is apparent that
`there are two potential
`types of screening approaches, each with different
`objectives that will result in different choices of
`models. Where a broad based. non-mechanism
`
`required, a disease—specific
`oriented screen is
`panel of xenografts with widely differing but
`biologically relevant phenotypes is likely to be
`optimal. Knowledge of the pattern of response to
`a series of established drugs, for each component
`of the panel, is required. Such a panel might be
`
`

`

`262
`
`R Clarke
`
`expected to contain both sensitive and resistant
`models (see Table 1 for examples included in one
`possible panel for breast cancer). For a mechan—
`ism or structure/function based screen, the choice
`
`of Components is likely to depend upon assump-
`tions inherent in the mechanism. For example,
`
`the panel
`where a specific target is identified,
`may contain several models with different levels
`of expression of the target, e.g., P-glycoprorein,
`multidrug resistance related protein [21,38,46,47].
`In many cases,
`this requirement may be most
`effectively met by a series of transfected cell lines
`and their respective control populations.
`
`Phenotypic stability
`
`The stability of the phenotype is a critical deter-
`minant for tumor model selection. Some tumor
`
`xenografts may require periodic Cycles of in vivo/
`in vitro growth in order to maintain the ease of
`reestablishment in vitro for some excision cyto-
`toxieity assays.
`Prolonged in viva growth of
`some established cell lines can re5ult in signif-
`icant phenotypic alterations. We have described
`the isolation of hormone-independent sublines of
`
`the estrogen-dependent MCF—7 human breast can-
`cer cell line following prolonged selection in vivo
`[17,18]. While reSponses to antiestrogens remain
`unaltered [18,48], there are significant changes in
`their responsiveness to estrogens [17,18,481 For
`
`many cell lines, this problem can be overcome by
`using cells within a limited number of passages
`(£10) from a single frozen stock of cells. The
`frozen stock should be from a single passage of
`cells with a well characterized phenotype.
`The intertumor stability of the growth patterns
`
`and cell cycle profiles also may be important
`considerations. A high degree of variability in
`intertumor growrh fractions could significantly
`influence the reliability or case of data interpre-
`tation. The stability of the metastatic potential
`must be well defined. Cells with an unpredictable
`
`metastatic capacity may alter tumor burden and
`affect survival and/or the host's sensitivity to the
`toxicity of cytotoxic treatments.
`
`Therapeutic index and dose scheduling
`
`The difference in the dose response curves of
`
`normal and neoplastic tissues, often referred to as
`the therapeutic window or therapeutic index,
`is
`widely applied in the clinical pharmacology of
`
`cytotoxic drugs [22,49-51]. This difference in
`drug sensitivity enables the administration of
`sufficient drug to produce cytotoxic effects in the
`tumor, but not to induce significant and irrevers-
`
`ible toxicity to normal cells. Unfortunately, many
`cytotoxic drugs exhibit a steep dose response
`curve with a small therapeutic index.
`In many
`cases, the development of unacceptable toxicity is
`the dose limiting factor for cytotoxic chemo-
`therapy.
`The sensitivity of normal cell populations
`
`reflects the rapid growth rate and high growth
`fraction of some cells.
`For example, cells in
`normal bone marrow and the intestinal crypts
`have respective thymidine labeling indices of
`30%—70% and 12%-18% [15]. These values are
`generally higher than observed in most solid
`tumor cell populations. This accounts for the
`high incidence of hematopoietic and gastro-
`intestinal
`side effects
`associated with many
`
`chemotherapeutic regimens. Granulocytes are
`highly susceptible to cytotoxic agents because of
`their high growth fraction and short lifespan, but
`a pool of non-cycling stem cells ensure repopula—
`tion [15]. Generally, the purpose of dose schedul-
`ing is to administer the second and subsequent
`treatments at times that will allow some normal
`
`stem cells to evade the drug and enable repopula—
`tion to occur without permanent damage to bone
`marrow. Examples of some drug schedules for
`rodents are provided in Table 2.
`
`Drug administration in vivo
`
`The route of administration can influence drug
`pharmacokinetics, toxicity. and antitumor activity.
`
`introduces the drug
`injection iv.
`For example,
`directly into the blood. with a relatively rapid
`exposure of hematopoietic stem cells. Adminis-
`
`

`

`tration so. or i.[9. would be expected to produce
`a slower drug equilibration with this compart-
`ment, and produce a delayed or reduced myelo~
`
`suppression. However, local tissue damage could
`be increased relative to the iv.
`route.
`The
`
`toxic/pharmacokinetic differences as—
`potential
`sociated with routes of administration can be
`
`For example,
`controlled by careful planning.
`While an i.v. bolus of an agent may produce
`
`the same dose in mg/kg
`unacceptable toxicity,
`body weight can occasionally be administered
`either by infusion, s.c,. 19.0., or multiple lower
`doses given i.v., with significant alterations in
`host toxicity.
`For the majority of solid tumors, an iv.
`administration most closely reflects the clinical
`administration of the drug. However, for experi—
`mental drugs, the route of administration will vary
`primarily with the physico—chemical properties of
`the drug. The iv. route is generally limited to
`water soluble compounds with a pH >4.0 and
`<85. Water~insoluble drugs can be administered
`either S.c., provided they do not produce un-
`acceptable local damage, or p.0.
`if they have
`sufficient chemical stability.
`For steroid hor—
`rnones and untillorinones that can require S115“
`
`tained delivery, i.p. or 3.0. depots in peanut oil,
`S. c. Alzet mini pumps (Alza Scientific, Palo Alto
`CA), cholesterol—based slow release pellets (In—
`novative Research of America, Sarasota, FL) or
`
`silastic pellets all can produce appropriate plasma
`levels of drug for sustained periods of time.
`The timing of administration also is an im-
`portant consideration.
`Initiation of treatment
`within a few days of tumor cell inoculation may
`produce evidence of activity, when administration
`to established tumors indicates inactivity, For
`
`cytotoxic agents, administration within a few days
`of cell inoculation is often inappropriate. Treat-
`mcnt of established tumors, where these are
`
`clearly palpable and fall within a predefined size
`range,
`is generally more appropriate and allows
`for assessment of the most widely used endpoints.
`
`This size should not be so large as to influence
`response to the drug. Early administration of
`drug is usually appropriate when the tumor is
`
`Breast cancer models for drug screenng
`
`263
`
`directly xenografted from another animal and has
`a rapid TD of only a few days, or perhaps when
`sensitivity is
`required in a primary screen.
`Another exception is for the testing of chemo-
`preventive agents, cg. antiestrogens or retinoids,
`which could be given to high risk women without
`evidence of clinically detectable disease. With
`these compounds, chemopreventiveichemosup-
`pressive activity against low tumor burde

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