throbber
British Journal of Cancer (1999) 79(11/12), 1633—1640
`© 1999 Cancer Research Campaign
`Article no. bjoc.1998.0261
`
`Review
`
`Making the most of rodent tumour systems in cancer
`drug discovery
`
`MC Bibby
`
`Clinical Oncology Unit, University of Bradford, Richmond Road, Bradford, West Yorkshire. BD7 1DP, UK
`
`It is clear that even after almost halfa century of intensive effort to
`develop effective treatments for common solid cancers there is
`still seine way to go before a major impact on survival of patients
`with these malignancies is achieved. Much of the paucity of
`success is blamed on the lack of appropriate models and there is a
`commonly held belief amongst cancer researchers that
`trans—
`plantable tumours in rodents are sensitive to drug therapy, are easy
`to cure and therefore not predictive of responses in humans. it is
`true that,
`in the past, when one considers the large number of
`compounds evaluated, murine tumour models have identified only
`a limited number of clinically useful agents and not a single
`cancer—specific drug has resulted from a murine tumour screen. It
`is easy with hindsight to blame this disappointing lack of really
`effective therapies on inappropriate test systems and screening
`strategies but, although drug discovery from rodent systems has
`been sparse, in reality, an enormous contribution to cancer therapy
`principles has emanated from such studies. This work really began
`in the mid 1950s when the National Cancer Institute (NCI) in the
`USA initiated a large—scale anti~cancer drug—screening programme
`using three murine
`tumour models:
`sarcoma
`180, L1210
`leukaemia and carcinoma 755, (Plowman et al. 1997). Of major
`significance in the early days was the work of Skipper and
`colleagues who, by the use of experimental
`leukaemia models,
`established the principle of
`survival/inoculum relationship
`(Skipper ct a1, 1957) and also the concept offractional cell kill, i.e.
`that the percentage ofthe tumour cell population killed by a given
`dose of drug was relatively constant (Skipper et a1, 1964). They
`also described a close relationship between dose level and
`percentage
`of
`leukaemic
`cell
`population
`killed,
`i.e.
`a
`doscmrcsponse relationship. Other useful
`therapeutic strategies
`were first established in rodent systems. The principle of optimum
`scheduling was first described by Goldin et a1 (1956) using the
`L1210 leukaemia. Combination chemotherapy and the concept of
`therapeutic synergism was first
`investigated in rodent models
`(Goldin and Mantel, 1957) as was combining the modalities of
`surgery and chemotherapy
`adjuvant chemotherapy (Martin,
`1981).
`studies utilized murine
`the early experimental
`Many of
`leukaemias which grew very rapidly, had a high growth fraction
`and proved to be sensitive to a number of agents that were subse—
`quently shown to have more activity against
`leukaemias and
`lymphomas than against solid carcinomas or sarcomas and to be
`toxic to the bone marrow (Muggia, 1987). A number ofinvestiga—
`
`Received 10 July 1998
`Revised 24 August 1998
`Accepted 16 September 1998
`
`Correspondence to: MC Bibby
`
`tors believed that the use of more appropriate, slower growing
`tumour models of solid cancers. and adoption ofclinically relevant
`end-points, would improve the usefulness of preclinical studies,
`particularly for agents with potential activity against the common
`solid cancers. Corbett et a1 (1987) pointed out that most of the
`agents that had entered the clinic at that time had poor, or no,
`activity against the majority of transplantable solid tumours in
`mice but they also suggested that, as negative findings are rarely
`reported, the casual reader of the drug discovery literature may
`have gained the inaccurate impression that transplantable tumours
`in mice are highly vulnerable to a large proportion of agents that
`make the clinic.
`
`THERAPEUTIC INDEX
`
`The importance of addressing the therapeutic index of investiga—
`tional agents has been stressed by many groups over the years,
`including our own (Double and Bibby, 1989) and we have advo—
`cated a more thorough preclinical evaluation of new cytotoxic
`entities which assesses efficacy and normal tissue toxicity in the
`same setting (Bibby ct a1, 1988). Taking mitozolomide (Stevens
`et a1. 1984) as an example compound that had recently entered
`clinical trial, we evaluated anti-tumour activity in a limited panel
`of refractory murine tumours but we also investigated normal
`tissue toxicity at the active doses. Mitozolomide showed a poor
`therapeutic ratio in this test system with modest anti—tumour
`effects being seen close to maximum tolerated dose (MTD) only.
`At this dose level there was very severe normal tissue toxicity in
`mice (Table l). Mitozolomide is an example of (at that time) a
`novel chemical that showed exciting pre—clinical activity in some
`experimental tumour systems with cures in L1210 and P388 at half
`the LDlO but went on to behave poorly in the clinic due to unman-
`ageable haematological toxicity. Although rodent studies do not
`
`Table 1 Bone marrow toxicity of mitozolomide assessed by spleen colony
`forming unit (CFU-S) assay in mice
`
`
`Donor
`Recipient
`Dose
`Radiation
`No. of bone marrow cells
`CFU-S
`
`(mg kg")
`(Gy)
`injected
`
`»~
`20
`30
`40
`
`11.7
`11.7
`11.7
`11.7
`
`1.2 x 10"
`1.0 x 105
`9.0 x 10“
`1.0 x 105
`
`4O
`4
`3
`0
`
`n = 6 mice per group. Data taken from Bibby et a1 1988.
`
`Genentech 2075
`Celltrion v. Genentech
`|PR2017—01122
`
`1633
`
`Genentech 2075
`Celltrion v. Genentech
`IPR2017-01122
`
`

`

`1634 MC Bibby
`
`necessarily predict human organ toxicity, they do appear to be
`helpful
`in predicting effects in some organs. e.g. bone marrow.
`Hindsight has shown that mitozolomide was selected for clinical
`trial on the basis of results from inappropriate pie—clinical models
`that did not reflect the resistance of clinical disease, but the group
`has gone
`on
`to develop
`a useful
`less
`toxic
`analogue,
`Temozolomide (Newlands et al, 1997).
`
`THERAPEUTIC END-POINTS
`
`In the early 19805, Martin and colleagues discussed the signifi—
`cance ofthe methodology used in recording response (Martin et a1,
`1984). They assessed the importance of selecting an appropriate
`end—point
`in preclinical studies by comparing response rates in
`breast cancer patients to six agents and then evaluating the same
`six agents in a murine model of spontaneous breast cancer. the
`CD8F1 (Stolfi et a1, 1988). The breast cancer patients responded
`only to melphalan, cyclophosphamide and 5—iluorouracil (S-FU).
`When the authors evaluated response in the murine system by
`percentage tumour inhibition, all six agents could be classified as
`active, whereas when they employed tumour regression as a
`measure of activity, as in the patients, only the three clinically
`active agents were effective (Table 2). Despite these observations
`cytotoxic agents have still progressed to clinical study on the basis
`of tumour growth delay in rodents at lVlTD and have often been
`shown to be ineffective in humans at doses that can be tolerated.
`Obviously there are certain ethical considerations that need to
`be addressed when selecting model systems and end-points for in
`vivo studies. As long as appropriate guidelines for animal welfare
`are followed (e.g. UKCCCR; Workman et al, 1998), subcutaneous
`models of neoplasia are relatively straightforward to use and
`potential animal suffering can be monitored and appropriate
`humane end-points, such as tumour size, utilized. However, the
`use of ascites tumour models and survival end—points to investi-
`gate the activity of intraperitoneally administered agents is diffi-
`cult to justify on scientific or ethical grounds. These should not be
`used for preclinical screening as they can result in undue suffering
`and provide little more information than well-designed in vitro
`assays. Lethality end-points should be avoided in general. even for
`models of systemic disease, and histological and/or metastatic
`colony counting techniques should be employed to assess anti—
`tumour effects. As modern therapeutic strategies develop, more
`
`effort is required to ensure suitable pharmaeodynamic end—points
`are in place to assess whether or not a designed therapy is
`achieving its goal in vivo.
`
`IN VITRO CELL LINE PANELS
`
`As a result of expanding knowledge, but also limited success, in
`the identification and development of drugs with useful activity in
`common solid cancers, drug—screening programmes in the NCI
`and elsewhere have
`evolved considerably over
`the years
`(Plowman et a1, 1997) until in 1985 the NCI began to assess the
`feasibility of using human tumour cell
`lines for
`large—scale
`compound screening (Boyd, 1989). It is beyond the scope of this
`article to extol the virtues or otherwise of this approach to drug
`discovery but it is clear that, as a result ofits inception, many inter-
`esting molecules are continually emerging and these will require
`appropriate in vivo evaluation. The NCI cell
`line panel has
`expanded since it was initially set up and there is ongoing develop-
`ment and characterization of potential new molecular targets.
`Although the initial strategy for the cell line screen was to develop
`a disease—orientated approach, the addition of the molecular char~
`acterization of the cell
`line panel opens up the possibility for
`target-orientated screening. The option to use genetically modified
`cell lines to address specific molecular targets at the in vitro stage
`is now available. This scientifically sound strategy oftarget-orien-
`tated drug development must be extended into in vivo evaluation
`protocols, so not only should in vivo studies be carried out
`in
`tumours derived from cell lines that were identified as sensitive in
`
`vitro, but it is imperative that adequate characterization of these
`models demonstrates that the molecular characteristics of the cell
`
`line are retained in the in vivo setting.
`
`PHARMACOKINETIC CONSIDERATIONS
`
`Going from cell—free screening strategies and cell lines to in vivo
`studies represents a major step in the drug—discovery pathway and
`one very important requirement is knowledge of the pharmaco—
`kinetics of investigational agents at an early stage. ideally, such
`studies should be carried out alongside efficacy investigations
`with a view to either selecting those agents with the best pharma-
`ceutical properties or, alternatively, formulating or even modifying
`the active component to optimize the therapeutic effect.
`
`
`
`Table 2 importance of clinically relevant endpoints in predictability of preclinical anti-tumour studies
`Drug
`Human
`CD8F1 spontaneous
`breast cancer
`breast tumours
`
` inhibited (%)a Assessment Regression Assessmentb
`
`
`
`
`
`
`
`Meiphalan
`Cycio
`FU
`
`+
`+
`+
`
`88
`80
`79
`
`+
`+
`+
`
`7/24
`5/25
`13/60
`
`+
`+
`+
`
`N-phosphon-acetylaspartate
`—
`63
`+
`2/47
`~
`Ara-c
`—
`33
`+
`1/37

`— 52 + 0/456‘thioguanine —
`
`
`
`
`
`
`
`
`
`
`
`aStatistically significant; bat least 50% reduction. Data taken from Stoifi et a1 1988.
`
`British Journal of Cancer (1999) 79(11/12), 7633—1640
`
`© Cancer Research Campaign 1999
`
`

`

`HOLLOW-FIBRE ASSAYS
`
`One relatively new approach to in vivo drug testing has been
`developed at NC] utilizing human cell-lines growing in hollow
`fibres. It is intended as a method for prioritizing compounds for
`testing in xenografts. At present. 10 000 compounds are screened
`in vitro against the cell line panel each year by NCl and 840% are
`referred for in vivo testing (Plowman et al, 1997). For the hollow—
`fibre assay,
`tumour cells are inoculated into l—mm internal
`diameter hollow—fibres that are heat—sealed and cut
`into 2cm
`
`lengths. The fibres are maintained in in vitro culture for 24—48 h
`and then implanted intraperitoneally and subcutaneously into nude
`mice. Three different cell lines can be grown in two different sites
`in the same mouse. The effects of treatment are determined by
`MTT assay on removal of the fibres 6~~8 days post—implantation.
`On the face of it, this technique would seem an efficient way
`of identifying lead compounds of promise because it requires
`relatively small expenditure and a
`limited quantity of test
`compound. Of course, as mentioned earlier, it cannot be assumed
`that expression ofa particular target will be identical in vivo to that
`expressed when the same cells are grown in vitro or vice versa.
`
`TUMOUR CHARACTERIZATION
`
`For several years our laboratory has been involved in studies of
`potential bioreductive drugs but in particular we have developed
`an interest in the role of the dimeric flavoprotein DT—diaphorase
`(DTD, NAD(_P)H:Quinone acceptor reductase, E.C.1.6.99.2) in the
`activation of a number of quinone based anti-cancer drugs
`(Phillips, 1996). There is a wealth of literature indicating a good
`correlation between enzyme activity in cell
`lines and aerobic
`chemosensitivity to compounds like E09. 3‘hydroxy—S-aziridinyl—
`l—methyl—2( 1 H-indole-4,7-dione)-prop-B-en—0t—ol
`(Robertson et
`al, 1992; Smitskamp-Wilms et al, 1994: Collard et al, 1995;
`Fitzsimmons et al, 1996). However, when attempts were made to
`translate these in vitro observations into in vivo studies it became
`
`apparent that DTD activity in human tumour xenografts derived
`from cell lines did not usually mirror levels seen in vitro (Collard
`et al, 1995),
`indicating the necessity to further characterize
`xenografts for the appropriate target for which therapeutic mole-
`cules are being sought.
`In the case of DTD, antibodies are
`available (Segura—Aguilar et al, 1994) so it is possible to identify
`the precise location of the protein within tissue sections (Segura—
`Aguilar et al, 1994; Phillips et al, 1998), and similar immunolocal—
`ization can be carried out now with a whole host of target enzymes
`and other molecular targets. It is certain that the majority oftargcts
`currently being investigated for drug development strategies will
`be influenced by their local environment, so not only will there be
`differential target expression in different tumours but there will
`also be different expression in vivo from that seen in in vitro
`culture of the same tumour cells. Furthermore,
`it
`is clear that
`tumour site within the body can influence the expression of
`specific targets, cg. P—glycoprotein. Fidler and colleagues investi-
`gated response to doxorubicin (DOX) or S—FU in three tumour
`types growing in different anatomical sites (Dong et al, 1994;
`Fidler et al, 1994). Sensitivity to 5~FU did not alter with anatom-
`ical site but
`lung and tumour deposits were resistant to DOX
`whereas subcutaneous (s.c.) tumours were sensitive. The authors
`determined that
`the difference in response was not due to
`differences in DOX distribution or potency but resulted from
`
`Rodent tumour systems in cancer drug discovery 1635
`
`over expression of mdrl mRNA in the resistant sites, they made
`the valid point that human colon cancer xenografts growing sub—
`cutaneously in nude mice often respond to DOX, whereas human
`colon cancer does not. These observations reiterate the require-
`ment for continual target characterization in in vivo models. Since
`modern molecular biology allows for the dissection of signalling
`pathways instrumental in cell proliferation and death, the number
`of potential molecular targets for drug development is increasing.
`Preclinical systems must be selected or designed in such a way to
`ensure that the appropriate target is expressed, and the appropriate
`controls in which the target is down—regulated or missing should
`be used in order to establish proof of principle and to ensure the
`validity of the therapeutic mechanism.
`
`HOST EFFECTS
`
`Although a molecular target approach using high—tlu‘oughput,
`robotic cell—free in vitro screens or cell
`line panels followed by
`properly characterized in vivo models should increase the effi—
`ciency of drug—discovery programmes, novel therapeutic strategies
`for cancer are clearly not going to be identified only by in vitro
`screening. These screens do not take into consideration indirect
`mechanisms such as host metabolism to an active species or
`immunomodulation. Within the rapidly expanding field oftumour
`biology several new therapeutic avenues are being explored that
`address host/tumour relationships or exploit specific features of
`solid tumours,
`thus relying even more on clinically relevant
`preclinical in vivo models.
`
`TU MOUR BLOOD SUPPLY
`
`One area of solid tumour biology that has been receiving a great
`deal of attention over recent years is the tumour blood supply.
`Broadly speaking,
`to date there are four different strategies
`attempting to exploit the tumour blood supply for therapeutic gain
`and all rely not only on differences in the architecture and cellular.
`biochemical and molecular properties between normal and tumour
`vasculature but also on the use of a properly characterized model.
`The major thrust at present seems to be concerned with anti-angio—
`genesis. i.e. a strategy to prevent the development of new blood
`vessels and to restrict solid tumour growth. There has been an
`enormous leap in our understanding of the processes and molec-
`ular control of tumour angiogenesis in recent years and the anti-
`angiogenic approach has been the subject of seine excellent
`comment articles and reviews (Folkman, 1990, 1997; Baillie et al,
`1995; Pluda, 1997). Modulation of tumour growth by the use of
`monoclonal antibodies to vascular endothelial growth factor
`(VEGF) has proved successful in preclinical studies (Warren et a1,
`1995) but the rational design of drugs which will specifically inter—
`fere with steps in the angiogenic process in tumours is in its
`infancy as is the development of gene therapy approaches (Kong
`and Crystal, 1998). However,
`targets are being identified. so
`continual development, improvement and characterization of in
`vivo tumour models in which to test specific strategies is required.
`The other attempts to exploit tumour blood supply have concen-
`trated on the existing blood vessels within solid tumours. One
`approach relies on the lack of smooth muscle and innervation of
`the new blood vessels growing in solid tumours. Several experi-
`mental studies have demonstrated potentiation of the activity of
`both standard and investigational drugs by combination with a
`
`@ Cancer Research Campaign 7 999
`
`British Journal of Cancer (1999) 79(11/12), 1633—1640
`
`

`

`1636 MC Bibby
`
`Normal tissue
`
`*
`
`9!
`
`’k
`Lymphatic drainage
`69
`\G)
`
`6),!
`
`it
`
`*
`it
`*
`it
`{37*
`ff :miteddrainage
`im‘fififl *
`
`‘k”:;€*7fir* G
`H: :12? 1:
`r
`*
`
`*
`
`at
`
`Continuous endothelium
`
`Leaky endothelium
`
`Figure 1
`
`Diagramatic representation of Enhanced Permeability and Retention in tumours (Matsumura and Maeda, 1986)
`
`
`
`i
`
`Glycine
`
`r
`i
`$HCH2‘Q
`90
`W CH3
`
`CH2
`
`l
`Phenylalanine
`i
`1
`Leucine
`
`
`Glycine
`
`
`
`Figure 2 Chemical structure of PK1 (Duncan, 1992)
`
`variety of vasoactive agents. A large number of agents alter blood
`flow in tumour models (Hirst and Wood, 1989) but many experi—
`mental studies utilized the anti-hypertcnsive hydralazine which
`has been shown to decrease blood flow through transplantable
`tumours in rodents (Chan et a1, 1984; Jirtle, 1988). Hydralazine
`enhanced the effectiveness of bioreductive drugs, such as the
`nitroimidazole RSU 1069 (Chaplin and Acker, 1987), tirapaza-
`mine (Brown, 1987), 1309 (Bibby et a1, 1993) and mitomycin C
`(Cowen et a1, 1994) but potentiation was also seen with melphalan
`
`(Stratford et a1, 1988) and tauromustine (Quinn et a1, 1992).
`Although these and other similar studies indicated a potential ther—
`apeutic strategy they have not yet resulted in a clinical advantage.
`Rowell ct a1 (1990) showed that single-dose oral hydralazine
`caused the blood flow through human lung tumours to decrease
`rather than increase. A study by Field et a1 (1991) demonstrated a
`difference in the effects of hydralazine on vascular perfusion
`between a series of primary and subcutaneously transplanted
`malignancies. The transplanted tumours responded to hydralazine
`
`British Journal of Cancer (1999) 79(17/72), 1633—7640
`
`© Cancer Research Campaign 1999
`
`

`

`whereas the primary tumour from which they were derived did not.
`Only 4/11 primary tumours responded to hydralazine. All
`the
`tumours used in the preceding papers were transplanted subcuta—
`neously in mice so, although they may have established proof ofa
`particular hypothesis,
`the studies are unlikely to be clinically
`predictive. An investigation from this laboratory (Cowen et al,
`1995) demonstrated that hydralazine was more effective at shut—
`ting down blood supply to so murine colon tumours than to the
`same turnouts transplanted orthotopically, and the same dose of
`hydralazine was completely ineffective in liver metastasis in this
`model. These studies highlight
`the lack of prediction by s.c.
`tumours for this strategy and the importance of selecting clinically
`relevant tumour models in appropriate anatomical sites.
`Another approach to targeting solid tumours by means of their
`poorly formed blood vessels relies on the enhanced permeability
`and retention effect (EPR) coined by Matsumura and Maeda
`(1986) (Figure 1). Many experimental tumours appear to selec-
`tively concentrate macromolecules of around 20 kDa molecular
`mass. This phenomenon is not due to just the leakiness of the
`tumour vasculature but also lack of the organized lymphatic
`drainage that usually occurs in normal tissues. One strategy that
`seems to be clinically useful at present is the incorporation of cyto-
`toxic moieties into copolymers of hydroxypropylmethylacryl—
`amide (HPMA) (Duncan, 1992). One ofthese compounds, PKl, is
`a copolymer with pcptidyl side—chains terminating in DOX (Figure
`2). The stable side—chain is thought to ensure that there is no extra-
`cellular release of DOX so that normal tissue toxicity is reduced.
`The polyrnerwdrug conjugate was designed so that
`the intact
`rnacromolecule is taken up into cells by pinocytosis and trans~
`ferred to the lysosomal compartment where it
`is exposed to an
`array of enzymes resulting in release of DOX. PKl containing at
`least four times the normal clinical dose of DOX was delivered to
`
`patients in a phase I trial and two responses were seen (Connors
`and Finedo, 1997). This kind of targeting seems a worthwhile
`strategy to pursue, but
`it
`is imperative that appropriate in vivo
`tumour models are employed in order to ensure the correct mole-
`cules are selected rapidly for clinical study. Preclinical studies with
`PKl
`indicated that, not only was it active against vascularized
`solid tumours in mice, it was also active against L1210 leukaemia
`when delivered by the intraperitoneal route (Duncan et al, 1992).
`Recent studies in this laboratory have also demonstrated that
`tumours in which the blood vessels are not leaky are no more
`responsive to PKl
`than to DOX. This study also demonstrated
`activity against avascular murine colon cancer
`lung colonies
`(Loadman et al, 1998) suggesting that, although there is impelling
`evidence from numerous studies that EPR plays a major role in the
`activity of PKl,
`the absence of such a phenomenon from
`rnicrornetastases would not preclude activity. Appropriate studies
`are required to further investigate this interesting approach.
`There is evidence emerging that
`the endothelium of solid
`tumour vasculature itself may be a useful target for drug therapy. It
`has been known since the 19305 that colchicine caused anti-
`
`vascular effects in experimental tumours (Clearkin, 1937) and it is
`now clear that some clinically useful tubulin interactive agents
`possess a vascular component in their mechanism of action against
`murine solid cancer models (Hill et al, 1993). However,
`these
`effects are seen only at doses close to MTD; the therapeutic index
`is extremely small so the ratio of sensitivity of tumour endothe~
`lium to normal cells is also small. A number of stilbenes,
`the
`cornbretastatins, has been isolated from the South African bush
`
`Rodent tumour systems in cancer drug discovery 1637
`
`willow, Combrerum crqffrum, and based on them a series of
`synthetic analogues has been synthesized (Pettit et al, 1989, 1995).
`Cornbretastatin A4 has been shown to bind to tubulin at
`the
`colchicine—binding site (McGown and Fox, 1989) and cause
`tumour blood flow reduction (Chaplin ct al, 1996), and Dark et al
`(1997) have demonstrated vascular shutdown with combretastatin
`A4 phosphate prodrug in so. experimental tumours at less than
`one—tenth of the MTD. Fearing the possibility of transplantation
`artefacts in these anti—vascular effects we have examined the
`
`efficacy of both combretastatin A4 and its prodrug on orthotopi—
`cally transplanted rnurine colon tumours and ensuing metastatic
`
`’D1o
`
`3
`'2{as}
`
`
`
`Figure 3 Haemorrhagic necrosis in an orthotopically implanted colon
`tumour and its metastasis following treatment with the investigational agent
`combretastatin A4 (courtesy of K Grosios). (A) Control caecal implant;
`(B) treated caecal implant; (C) treated metastatic deposit
`
`© Cancer Research Campaign 1999
`
`British Journal of Cancer (1999) 79(11/12), 1633—1640
`
`

`

`1638 Me Bibby
`
`deposits (Grosios et a1, 1997). Both parent compound and prodrug
`caused major haemorrhagic necrosis in tumours at the orthotopic
`site and in metastatic deposits (Figure 3). Careful examination of
`morphological effects indicated that
`lung deposits that had not
`undergone neovascularization did not respond to either compound,
`indicating that vaseularization and not
`transplantation site
`seems to be the important component for the activity of these
`compounds. The cornbretastatin A4 prodrug is awaiting phase 1
`clinical trial and it remains to be seen whether the vascular effects
`
`predicted from the preclinical models will occur.
`Another compound selected for clinical
`trial on the basis of
`preclinical anti—vascular effects is 5,6-dimethyl xanthenone acetic
`acid (XAA). This is a more potent analogue of flavone acetic acid
`(FAA) (Atassi et a1. 1985) from a series developed by Baguley and
`colleagues in Auckland, New Zealand (Atwell et
`a1, 1989;
`Rewcastle et a1, 1989, 1991a, 1991b, 1991c). FAA is an interesting
`molecule that had impressive experimental activity but, disap—
`pointingly, failed to show any clinical activity (reviewed by Bibby,
`1991; Bibby and Double, 1993). XAA again showed impressive
`haemorrhagic necrosis in vascularized solid tumour models but
`this time at tenfold lower doses than FAA (Rewcastle et al, 1991a:
`Ching et a1, 1992; Laws et al, 1995a) and also caused widespread
`haemorrhagic necrosis in an orthotopic model of human colon
`cancer (Laws et al, 1995b). Phase 1 clinical trials are ongoing in
`Auckland and in Mt Vernon Hospital/Gray Laboratory and
`Bradford, UK. To date, tumour vascular effects have been noted in
`two patients (Rustin et a1, 1998) suggesting that the preclinical
`models may well have been predictive in this case but, more
`importantly, demonstrating proof of principle, i.e. that a vascular
`effect demonstrated in in vivo preclinical models can also occur in
`solid malignancies in man. These clinical effects suggest that this
`anti—vascular strategy may well be worth pursuing with other more
`potent chemical entities.
`A particularly interesting development that again relies on thor-
`ough characterization of in vivo models has come from the identi—
`fication that certain peptides home specifically to the vasculature
`ofspecific organs (Pasqualini and Ruoslahti, 1996) and to tumour
`vasculature (Arap et a1, 1998). In the latter study the authors have
`demonstrated that it may be possible to target chemotherapeutic
`drugs specifically to tumour vasculature on the basis of differential
`expression of receptors.
`
`ORTHOTOPIC TUMOUR MODELS
`
`It is now clear from large numbers of studies, some of which are
`cited here, that it is necessary to take account of not only the rele-
`vanee of the tumour type utilized for the in vivo evaluation of
`novel drugs and other therapeutic strategies but also tumour site.
`In order to do this it is necessary to use model systems that reflect
`the morphology and growth characteristics of clinical disease but,
`equally as importantly, must be technically feasible and ethically
`acceptable to carry out. For the last 20 years good studies have
`been reported in the literature indicating that such orthotopic
`systems exist (eg. Tan et a1, 1977; Goldrosen et a1, 1986; Bresalier
`et a1, 1987; Morikawa et a1, 1988; Fu et a1, 1991) and the worth of
`such systems has been extensively reviewed (Fidler et a1, 1990;
`Fidler, 1991; Gutman and Fidler, 1995; Hoffman, 1997).
`It
`is
`important to remember that, in the case of common solid cancers
`in humans, the primary tumour mass is usually excised by the
`surgeon and it
`is the metastases that require novel
`therapeutic
`
`strategies, so models should reflect these clinical metastases. It is
`still necessary to refine orthotopic techniques in order to improve
`the predictability of these models in the future, either by character—
`ization for specific molecular targets within these clinically rele—
`vant sites of metastasis, or to develop similar models with tumour
`cells appropriately transfected to express the target of interest. It
`must be remembered that, if one adopts an orthotopic approach
`with models for drug discovery, due attention is given to appro-
`priate end-points. Lethality should not be seen as an alternative to
`a specific pharmacodynamic end—point such as changes in blood
`flow in the case of anti-vascular strategies or production of
`cytokines by immunornodulators etc.
`
`CONCLUSIONS
`
`It is clear that murine tumour models have been very helpful in
`determining basic principles of cancer chemotherapy and to date
`have been instrumental
`in identifying and evaluating a limited
`number of clinically useful agents, Contrary to popular belief
`many murine tumours are not easy to cure with standard
`chemotherapeutic agents and, ifadequate attention is paid to clini-
`cally relevant end—points and therapeutic index, they can be good
`predictors of clinical activity. Of course today a whole host of
`approaches and assays is available to drug—discovery teams:
`
`-
`-
`
`'
`-
`
`high throughput robotic cell-free screens
`in vitro cell lines (sometimes genetically modified to express a
`specific target)
`hollow—fibre in vivo assays
`induced rodent tumour systems W usually intraperitoneal or
`subcutaneous
`
`spontaneous rodent tumours
`-
`- models ofmetastasis
`
`-
`-
`-
`
`orthotopic models
`human tumour xenografts
`transgenic systems.
`
`Modern drug discovery should be much more mechanistically
`based and target-driven, so the first two of these systems should
`continue to lead to identification of structures that have potential
`for development. Assays like the NCI hollow-fibre screen provide
`a rapid and relatively inexpensive method for assessing in vivo
`potential. The remaining systems should be carefully selected
`based on the particular therapeutic approach. The evidence from
`the literature is clear in that it indicates that rodent tumours should
`
`not be used for random screening. lt is necessary to fully charac-
`terize tumour models to ensure that
`the precise target for the
`strategy being evaluated is expressed in vivo. Since targets are
`known to vary with different anatomical sites, more effort should
`be placed on developing models in clinically relevant sites rather
`than the continuous use of subcutaneously transplanted tumours
`whilst ignoring their tissue of origin or likely site of metastasis in
`humans. Ultimately, regardless ofthe molecule being investigated
`and developed,
`it
`is necessary to produce a safe pharmaceutical
`preparation that can be delivered in an appropriate way to reach
`the target for which it has been designed, so to make real progress
`in drug development appropriate in vivo rodent models are still
`essential. However,
`in addition to scientific considerations, due
`ethical consideration must be given to ensure selection of the
`appropriate model for the task in hand and animal welfare guide—
`lines rnust be followed throughout.
`
`British Journal of Cancer (1999) 79(11/12), 1633—1640
`
`© Cancer Research Campaign 1999
`
`

`

`In drug—discovery programmes it is not financially viable, nor
`does it make scientific sense,
`to use large panels of murine’
`tumours or human tumour xenografts
`for extensive in vivo
`screening, particularly when these xenografts are often poorly
`characterized. The most efficient and ethical approach must be
`to design limited, but
`relevant, experiments to address key
`questions, cg:
`
`- Can effective drug concentrations based on previous in vitro
`data be achieved in vivo?
`
`'
`
`'
`
`Is the molecule reaching its designated clinically relevant
`target?
`Is there efficacy or, in other words, is there proof of principle?
`
`If these questions can be answered in the affirmative then the
`compound should be scheduled for clinical trial without the need
`for further extensive in vivo testing.
`In summary,
`there are certain key requirements for in vivo
`studies:
`
`‘ Models must have known clinically-relevant target status.
`- Appropriate bioavailability oftest molecules should be demon-
`strat

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