throbber
(cid:5)(cid:6)(cid:7)(cid:8)(cid:9)(cid:10)(cid:11)(cid:3)(cid:6)(cid:12)(cid:3)(cid:13)(cid:14)(cid:15)(cid:16)(cid:6)(cid:17)(cid:14)(cid:15)(cid:8)(cid:10)(cid:18)(cid:19)
`
`(cid:20)(cid:21)(cid:21)(cid:22)(cid:23)(cid:3)(cid:24)(cid:24)(cid:25)(cid:26)(cid:4)(cid:26)(cid:26)(cid:27)(cid:28)(cid:3)(cid:29)(cid:30)(cid:8)(cid:31)(cid:9)(cid:17)(cid:32)(cid:3)(cid:24)(cid:27)(cid:33)(cid:34)(cid:4)(cid:27)(cid:35)(cid:33)(cid:36)(cid:3)(cid:29)(cid:37)(cid:9)(cid:11)(cid:31)(cid:9)(cid:15)(cid:32)(cid:3)(cid:5)(cid:6)(cid:7)(cid:8)(cid:9)(cid:10)(cid:11)(cid:3)(cid:14)(cid:6)(cid:16)(cid:15)(cid:18)(cid:10)(cid:38)(cid:15)(cid:23)(cid:3)(cid:14)(cid:17)(cid:17)(cid:18)(cid:23)(cid:39)(cid:39)(cid:40)(cid:40)(cid:40)(cid:41)(cid:17)(cid:10)(cid:9)(cid:42)(cid:12)(cid:6)(cid:9)(cid:11)(cid:31)(cid:9)(cid:15)(cid:41)(cid:43)(cid:6)(cid:16)(cid:39)(cid:11)(cid:6)(cid:31)(cid:39)(cid:19)(cid:44)(cid:6)(cid:43)(cid:25)(cid:26)
`
`(cid:45)(cid:15)(cid:46)(cid:15)(cid:8)(cid:47)(cid:31)(cid:9)(cid:38)(cid:3)(cid:45)(cid:15)(cid:47)(cid:31)(cid:47)(cid:17)(cid:10)(cid:9)(cid:43)(cid:15)(cid:3)(cid:17)(cid:6)(cid:3)(cid:48)(cid:10)(cid:8)(cid:38)(cid:15)(cid:17)(cid:15)(cid:42)(cid:3)(cid:48)(cid:14)(cid:15)(cid:8)(cid:10)(cid:18)(cid:19)
`
`(cid:49)(cid:41)(cid:3)(cid:50)(cid:31)(cid:42)(cid:10)(cid:11)(cid:51)(cid:3)(cid:52)(cid:41)(cid:3)(cid:53)(cid:17)(cid:17)(cid:10)(cid:8)(cid:42)(cid:51)(cid:3)(cid:21)(cid:41)(cid:3)(cid:54)(cid:10)(cid:19)(cid:15)(cid:3)(cid:55)(cid:3)(cid:5)(cid:41)(cid:3)(cid:56)(cid:15)(cid:3)(cid:57)(cid:6)(cid:9)(cid:6)
`
`(cid:48)(cid:6)(cid:3)(cid:43)(cid:31)(cid:17)(cid:15)(cid:3)(cid:17)(cid:14)(cid:31)(cid:47)(cid:3)(cid:10)(cid:8)(cid:17)(cid:31)(cid:43)(cid:11)(cid:15)(cid:23)(cid:3)(cid:5)(cid:6)(cid:3)(cid:7)(cid:8)(cid:9)(cid:10)(cid:11)(cid:12)(cid:3)(cid:13)(cid:6)(cid:3)(cid:14)(cid:15)(cid:15)(cid:10)(cid:16)(cid:9)(cid:12)(cid:3)(cid:17)(cid:6)(cid:3)(cid:18)(cid:10)(cid:19)(cid:20)(cid:3)(cid:21)(cid:3)(cid:22)(cid:6)(cid:3)(cid:23)(cid:20)(cid:3)(cid:24)(cid:25)(cid:26)(cid:25)(cid:3)(cid:27)(cid:28)(cid:29)(cid:29)(cid:30)(cid:31)(cid:3)(cid:32)(cid:20)(cid:33)(cid:20)(cid:16)(cid:34)(cid:8)(cid:26)(cid:35)(cid:3)(cid:32)(cid:20)(cid:34)(cid:8)(cid:34)(cid:15)(cid:10)(cid:26)(cid:36)(cid:20)
`(cid:15)(cid:25)(cid:3)(cid:37)(cid:10)(cid:16)(cid:35)(cid:20)(cid:15)(cid:20)(cid:9)(cid:3)(cid:37)(cid:38)(cid:20)(cid:16)(cid:10)(cid:39)(cid:19)(cid:12)(cid:3)(cid:22)(cid:25)(cid:40)(cid:16)(cid:26)(cid:10)(cid:11)(cid:3)(cid:25)(cid:41)(cid:3)(cid:42)(cid:38)(cid:20)(cid:43)(cid:25)(cid:15)(cid:38)(cid:20)(cid:16)(cid:10)(cid:39)(cid:19)(cid:12)(cid:3)(cid:44)(cid:45)(cid:46)(cid:34)(cid:40)(cid:39)(cid:30)(cid:12)(cid:3)(cid:47)(cid:4)(cid:44)(cid:28)(cid:12)(cid:3)(cid:23)(cid:48)(cid:49)(cid:46)(cid:3)(cid:44)(cid:29)(cid:6)(cid:44)(cid:44)(cid:47)(cid:50)(cid:51)
`(cid:52)(cid:25)(cid:36)(cid:6)(cid:28)(cid:29)(cid:29)(cid:30)(cid:6)(cid:44)(cid:45)(cid:6)(cid:17)(cid:40)(cid:39)(cid:39)(cid:11)(cid:20)(cid:43)(cid:20)(cid:26)(cid:15)(cid:4)(cid:44)(cid:6)(cid:47)
`
`(cid:48)(cid:6)(cid:3)(cid:11)(cid:31)(cid:9)(cid:58)(cid:3)(cid:17)(cid:6)(cid:3)(cid:17)(cid:14)(cid:31)(cid:47)(cid:3)(cid:10)(cid:8)(cid:17)(cid:31)(cid:43)(cid:11)(cid:15)(cid:23)(cid:3)(cid:3)(cid:38)(cid:15)(cid:15)(cid:39)(cid:46)(cid:51)(cid:51)(cid:9)(cid:53)(cid:6)(cid:9)(cid:25)(cid:8)(cid:6)(cid:25)(cid:16)(cid:35)(cid:51)(cid:44)(cid:29)(cid:6)(cid:44)(cid:44)(cid:47)(cid:50)(cid:51)(cid:52)(cid:25)(cid:36)(cid:6)(cid:28)(cid:29)(cid:29)(cid:30)(cid:6)(cid:44)(cid:45)(cid:6)(cid:17)(cid:40)(cid:39)(cid:39)(cid:11)(cid:20)(cid:43)(cid:20)(cid:26)(cid:15)(cid:4)(cid:44)(cid:6)(cid:47)
`
`(cid:54)(cid:40)(cid:24)(cid:11)(cid:8)(cid:34)(cid:38)(cid:20)(cid:9)(cid:3)(cid:25)(cid:26)(cid:11)(cid:8)(cid:26)(cid:20)(cid:46)(cid:3)(cid:44)(cid:55)(cid:3)(cid:23)(cid:20)(cid:36)(cid:3)(cid:28)(cid:29)(cid:44)(cid:55)(cid:6)
`
`(cid:17)(cid:40)(cid:24)(cid:43)(cid:8)(cid:15)(cid:3)(cid:19)(cid:25)(cid:40)(cid:16)(cid:3)(cid:10)(cid:16)(cid:15)(cid:8)(cid:36)(cid:11)(cid:20)(cid:3)(cid:15)(cid:25)(cid:3)(cid:15)(cid:38)(cid:8)(cid:34)(cid:3)(cid:52)(cid:25)(cid:40)(cid:16)(cid:26)(cid:10)(cid:11)(cid:3)
`
`(cid:14)(cid:16)(cid:15)(cid:8)(cid:36)(cid:11)(cid:20)(cid:3)(cid:33)(cid:8)(cid:20)(cid:56)(cid:34)(cid:46)(cid:3)(cid:44)(cid:44)
`
`(cid:7)(cid:8)(cid:20)(cid:56)(cid:3)(cid:16)(cid:20)(cid:11)(cid:10)(cid:15)(cid:20)(cid:9)(cid:3)(cid:10)(cid:16)(cid:15)(cid:8)(cid:36)(cid:11)(cid:20)(cid:34)(cid:3)
`
`(cid:42)(cid:8)(cid:15)(cid:8)(cid:26)(cid:35)(cid:3)(cid:10)(cid:16)(cid:15)(cid:8)(cid:36)(cid:11)(cid:20)(cid:34)(cid:46)(cid:3)(cid:57)(cid:3)(cid:7)(cid:8)(cid:20)(cid:56)(cid:3)(cid:36)(cid:8)(cid:15)(cid:8)(cid:26)(cid:35)(cid:3)(cid:10)(cid:16)(cid:15)(cid:8)(cid:36)(cid:11)(cid:20)(cid:34)(cid:3)
`
`(cid:58)(cid:40)(cid:11)(cid:11)(cid:3)(cid:37)(cid:20)(cid:16)(cid:43)(cid:34)(cid:3)(cid:21)(cid:3)(cid:42)(cid:25)(cid:26)(cid:9)(cid:8)(cid:15)(cid:8)(cid:25)(cid:26)(cid:34)(cid:3)(cid:25)(cid:41)(cid:3)(cid:10)(cid:36)(cid:36)(cid:20)(cid:34)(cid:34)(cid:3)(cid:10)(cid:26)(cid:9)(cid:3)(cid:40)(cid:34)(cid:20)(cid:3)(cid:36)(cid:10)(cid:26)(cid:3)(cid:24)(cid:20)(cid:3)(cid:41)(cid:25)(cid:40)(cid:26)(cid:9)(cid:3)(cid:10)(cid:15)
`(cid:38)(cid:15)(cid:15)(cid:39)(cid:46)(cid:51)(cid:51)(cid:56)(cid:56)(cid:56)(cid:6)(cid:15)(cid:10)(cid:26)(cid:9)(cid:41)(cid:25)(cid:26)(cid:11)(cid:8)(cid:26)(cid:20)(cid:6)(cid:36)(cid:25)(cid:43)(cid:51)(cid:10)(cid:36)(cid:15)(cid:8)(cid:25)(cid:26)(cid:51)(cid:52)(cid:25)(cid:40)(cid:16)(cid:26)(cid:10)(cid:11)(cid:49)(cid:26)(cid:41)(cid:25)(cid:16)(cid:43)(cid:10)(cid:15)(cid:8)(cid:25)(cid:26)(cid:59)(cid:52)(cid:25)(cid:40)(cid:16)(cid:26)(cid:10)(cid:11)(cid:42)(cid:25)(cid:9)(cid:20)(cid:60)(cid:19)(cid:52)(cid:25)(cid:36)(cid:28)(cid:29)
`
`Amerigen Exhibit 1147
`Amerigen v. Janssen IPR2016-00286
`
`(cid:56)(cid:6)(cid:40)(cid:9)(cid:11)(cid:6)(cid:10)(cid:42)(cid:3)(cid:57)(cid:19)(cid:23)(cid:3)(cid:61)(cid:62)(cid:16)(cid:3)(cid:63)(cid:8)(cid:11)(cid:11)(cid:8)(cid:10)(cid:43)(cid:3)(cid:64)(cid:10)(cid:16)(cid:20)(cid:65)
`
`(cid:56)(cid:10)(cid:17)(cid:15)(cid:23)(cid:3)(cid:44)(cid:28)(cid:3)(cid:22)(cid:10)(cid:26)(cid:40)(cid:10)(cid:16)(cid:19)(cid:3)(cid:28)(cid:29)(cid:44)(cid:47)(cid:12)(cid:3)(cid:14)(cid:15)(cid:46)(cid:3)(cid:44)(cid:44)(cid:46)(cid:66)(cid:30)
`
`

`
`REPRINT
`
`Journal of Chemotherapy
`
`Vol. 16 - Supplement n. 4 (7-12) - 2004
`
`REVIEW
`
`Reversing Resistance to Targeted Therapy
`
`L. VIDAL - G. ATTARD - S. KAYE - J. DE BONO*
`
`Institute of Cancer Research and Royal Marsden Hospital, Sutton, UK.
`
`* Correspondence: Dr. Johann S. de Bono, MB ChB, FRCP, MSc, PhD, Senior Lecturer and Consultant Medical
`Oncologist, Centre for Cancer Therapeutics, Institute for Cancer Research, Royal Marsden Hospital, Downs Road,
`Sutton, Surrey SM2 5PT, England. Tel: +44-20-8722-4028; Fax: +44-20-8642-7979. E-mail: jdebono@icr.ac.uk
`
`Summary
`
`The development of molecular targeted anticancer drugs is rapidly changing
`cancer therapeutics. However, drug resistance to these novel agents remains a real
`clinical concern. Reports now indicate that resistance to many of these molecular
`targeted agents - including hormone therapies, trastuzumab, imatinib, and gefitinib
`- occurs via common resistance mechanisms. These include 1) inadequate target
`blockade due to sub-optimal drug delivery; 2) altered target expression at the DNA
`(gene amplification), mRNA or protein level; 3) an altered target such as a mutated
`kinase domain; 4) modified target regulating proteins (e.g. altered expression of co-
`activators and/or co-repressors for nuclear steroid hormone receptors); 5) signal-
`ling by alternative proteins (functional redundancy) or different signalling pathways.
`It is envisioned that the molecular evaluation of clinical anticancer drug resistance,
`which requires the detailed study of pharmacokinetics, pharmacogenetics and phar-
`macodynamics, will allow the development of rational reversal strategies and impro-
`ved patient outcome.
`
`Key words: Targeted therapy, resistance.
`
`INTRODUCTION
`
`SPECIFIC MECHANISMS OF RESISTANCE
`
`Preferential cytotoxicity against malignant tissues
`remains tantamount to the Holy Grail in cancer
`therapeutics because this portends improved patient
`tolerance and quality of life and, importantly, the
`capacity to deliver combination therapy. Rationally
`designed and molecular target based anticancer
`agents are characterised by lower toxicity and wider
`therapeutic indices than traditional cytotoxic drugs.
`These agents may reverse or modulate chemothera-
`py resistance, enhance anti-tumour activity or main-
`tain tumour regression. This article reviews potential
`mechanisms of resistance to targeted therapies and
`suggests strategies to overcome this resistance,
`thereby maximizing anti-tumour effect and clinical
`benefit.
`
`Anti-androgen resistance
`
`Hormone therapy remains the mainstay for the
`treatment of metastatic prostate cancer. Androgen
`deprivation therapy induces a remission in 80 to
`90% of patients with advanced disease and results in
`a median progression-free survival of 12 to 33
`months, at which time an androgen-independent
`phenotype usually emerges. This accounts for the
`median overall survival of 23 to 37 months from the
`initiation of androgen deprivation 1. Various mecha-
`nisms of androgen resistance have been postulated.
`Inadequate LHRH and testicular androgen suppres-
`sion or circulating low levels of adrenal androgens
`can result in failure to respond to hormone therapy.
`Improving or increasing LHRH analogue delivery
`
`© E.S.I.F.T. srl - Firenze
`
`ISSN 1120-009X
`
`

`
`8
`
`L. VIDAL - G. ATTARD - S. KAYE - J. DE BONO
`
`can abrogate the former, while adrenal androgen
`synthesis can be inhibited by the administration of
`low doses of steroids or through the inhibition of
`key enzymes in the adrenal steroid biosynthesis
`pathways with agents such as ketoconazole or the
`CYP17 inhibitor abiraterone acetate 2. Preclinical
`data indicate that most androgen-independent
`prostate cancers continue to express androgen
`receptor (AR) with AR signalling remaining intact, as
`demonstrated by the expression of prostate-specific
`antigen (PSA), and consistently increased levels of
`AR mRNA, despite castrate testosterone levels 3.
`This could occur following binding of (low) levels of
`ligand to hypersensitive AR complexes or by andro-
`gen-independent activation of the AR. AR gene
`amplification resulting in increased receptor levels
`could also result in sensitisation of the cell to low lig-
`and concentrations. Similarly, point mutations of the
`AR gene may alter the antagonistic effect on the AR
`receptor seen with steroid ligands, such as oestro-
`gens, corticosteroids or even AR antagonists, to an
`agonistic one. Although these mutations appear to
`be uncommon in the clinic,4 they could explain the
`anti-androgen withdrawal syndrome observed in
`approximately 10-15% of hormone refractory
`prostate cancer (HRPC) patients 5.
`Hormone resistance in the majority of patients
`who do not have AR gene alterations but who retain
`active AR signalling may be explained by androgen-
`independent activation by growth factor signalling,
`which can directly activate the AR and its down-
`stream pathways. This may occur through activation
`of the erb-B receptors due to their aberrant expres-
`sion or the over-expression of their ligands 6. In
`addition, over-expression of insulin growth factor
`(IGF)-I and fibroblast growth factor (FGF)-8b has
`been reported in HRPC; binding to their respective
`receptors can bypass the AR and activate down-
`stream signalling 7,8. Ligand-independent activation
`of the AR can also occur through the increased
`expression of co-activators (such as SRC-1) or
`decreased expression of co-repressors (such as
`NCoR), which can alter AR transcriptional activity 9.
`Finally, alternative-signalling pathways involved in
`the regulation of apoptosis, such as the up-regula-
`tion of anti-apoptotic genes (e.g.Bcl-2 or the IAPs)
`could impart clinical androgen resistance. Strategies
`to reverse such resistance have been pursued in clin-
`ical trials utilising antisense technologies.
`
`ANTI-OESTROGEN RESISTANCE
`
`Intrinsic resistance or the development of
`acquired resistance to oestrogen manipulation
`remains a significant clinical problem in the treat-
`ment of oestrogen receptor (ER) positive breast can-
`cer 10. While up to 70% of patients with ER positive
`advanced disease initially respond to hormonal mod-
`ulation, most tumours will eventually become resis-
`
`tant. While the most important factor in defining
`hormonal resistance is lack of ER expression, the
`mechanisms of resistance in ER positive tumours
`have not been fully elucidated and may be multifac-
`torial.
`The ER pathway is complex and involves multi-
`ple co-regulating factors linked to other signalling
`pathways and in particular, is regulated through its
`phosphorylation on two serine residues which can
`be modulated by the PI3K/Akt and the Ras/Raf/Erk
`pathways. The ER forms multiprotein complexes
`with co-regulatory proteins that possess either his-
`tone acetylase or histone deacetylase activity. These
`co-regulators are recruited in the process of ER lig-
`and binding, increasing or blocking downstream
`effectors depending on the balance between co-acti-
`vators and co-repressors. Increased ER expression
`can alter the response of ER binding to ligands such
`as oestrogens or anti-oestrogens. Changes in host
`endocrinology, drug pharmacokinetics, altered α:β
`ER ratios, ER mutations, perturbations in down-
`stream growth factor signalling, and altered ER co-
`regulating factors have all been implicated in this
`resistance phenotype 10.
`Reports have described a mutation in the hor-
`mone-binding domain of ER-α that is hypersensitive
`to low levels of oestrogen compared with wild-type
`ER-alpha in preclinical models 11. This point muta-
`tion comprises an amino-acid substitution in the ER-
`α acetylation site 5 that alters the ability of the ER to
`bind to many co-regulatory factors and consequent-
`ly, its transcriptional activity. However, this mecha-
`nism of resistance remains controversial. Up-regula-
`tion of receptor co-activators is another possible
`mechanism of oestrogen resistance. Reports indicate
`that increased expression of co-activators may stimu-
`late growth in the absence of oestrogen 10. In this
`environment, tamoxifen may exert an agonistic
`rather than an antagonist effect. Furthermore, cross-
`talk between erb-B receptor signalling and the ER
`has been associated with endocrine therapy-resis-
`tance 10,12. The ER may be activated by phosphory-
`lation in the absence of its ligand oestradiol by
`growth factor signalling (e.g. EGF and IGF-1)
`through both mitogen-activated protein kinase and
`Akt pathways. Preclinical and clinical data have
`already demonstrated that epidermal growth factor
`receptor (EGFR) tyrosine kinase inhibitors may
`reverse resistance to tamoxifen, and clinical trials
`combining endocrine therapies and signal transduc-
`tion inhibitors are ongoing 12. Preliminary data from
`a phase II trial indicate that gefitinib is active in ER
`positive tamoxifen-resistant breast cancer patients
`13,14.
`Altered intracellular signalling, downstream of
`the receptor tyrosine kinases may also result in hor-
`mone resistance. This can be addressed by targeting
`downstream signal-transduction proteins that interact
`with the ER pathway; for example inhibiting Ras sig-
`nalling by the farnesyltransferase inhibitors (FTIs) or
`
`

`
`REVERSING RESISTANCE TO TARGETED THERAPY
`
`9
`
`the molecular target of rapamycin (mTOR) by the
`rapamycin analogues 15,16. Emerging clinical trial
`data indicate that these agents can reverse hormone
`resistance in advanced breast cancer.
`
`TRASTUZUMAB (HERCEPTIN™) RESISTANCE
`
`Many mechanisms may be involved in the devel-
`opment of resistance to trastuzumab. Redundant sig-
`nalling mediated by the hetero- or homo-dimeriza-
`tion of other members of the erb-B family (EGFR,
`HER3, HER4) may activate downstream signalling
`despite HER-2 blockade by trastuzumab 17.
`Trastuzumab resistance may be reversed by combin-
`ing agents that target different members of the erb-
`B family (for example, trastuzumab with gefitinib or
`trastuzumab with erlotinib) or using agents that tar-
`get more than one erb-B family member. Agents
`such as the human monoclonal antibody pertuzumab
`(Omnitarg/2C4) targeting the heterodimerization
`domain on HER2 or the pan-erbB small molecule
`inhibitors need to be evaluated in this setting.
`Trastuzumab resistance may also result from overex-
`pression of other growth factor receptors or their
`ligands (such as the IGF-1R) or altered downstream
`proteins (eg. loss of PTEN) that modulate down-
`stream signalling 18. Targeting downstream signalling
`with small molecule inhibitors such as the mTOR
`inhibitors, Raf kinase inhibitors or FTIs may there-
`fore reverse trastuzumab resistance. Finally, recent
`data suggest that some trastuzumab-resistant breast
`cancers express reduced p27kip1 levels with increased
`cdk2/4 activity, suggesting a role for drugs that
`induce p27kip1 protein expression or inhibit cdk2/4
`in the treatment of these patients 19. Small molecule
`cdk inhibitors are being evaluated in the clinic and
`may merit further evaluation in this setting.
`
`GEFITINIB RESISTANCE
`
`Gefinitib (IressaTM) is an EGFR tyrosine kinase
`inhibitor. Recent reports indicate that incomplete
`target blockade due to poor drug delivery may be a
`significant concern in the treatment of colorectal
`cancer 20. Tumour biopsies from 28 colorectal can-
`cer patients were collected before and after treat-
`ment with gefitinib (single agent gefitinib 250 mg vs
`500 mg on an ECOG randomised phase II trial
`comparing the two doses given continuously daily).
`EGFR, p-EGFR (tyrosine residue at position 1068),
`Akt, p-Akt, MAPK, p-MAPK and Ki67 were charac-
`terized 20. Only 3 tumours demonstrated a fall in p-
`EGFR after one week of gefitinib therapy while the
`remaining biopsies did not show p-EGFR inhibition
`indicating that gefitinib was not effectively inhibiting
`its target. No inhibition of downstream signalling or
`Ki67 expression was observed. In this study only 1
`of 110 patients treated had an objective response.
`
`Other reports indicate that patients who develop an
`acneiform skin rash while on treatment with an
`EGFR-targeting drug are more likely to show clinical
`benefit. This was noted in studies of patients with
`non-small cell lung cancer (NSCLC) treated with
`erlotinib and patients with colorectal carcinoma
`treated with cetuximab (ErbituxTM) 21. The develop-
`ment of skin rash reflects target blockade, suggesting
`that patients with higher drug exposure are more
`likely to respond. In an attempt to maximise target
`blockade and minimise inter-patient pharmacody-
`namic variability, it has been proposed that dosing
`with these agents should be increased until a grade
`1 or 2 (CTCAE) skin rash is observed.
`Incomplete target blockade is only one of several
`potential resistance mechanisms to treatment with
`EGFR inhibitors. Increased expression of the target
`EGFR or its ligands due to gene amplification,
`altered RNA stability or altered recycling can all
`result in gefitinib resistance. Mutations of the EGFR
`kinase domain can also result in altered EGFR sensi-
`tivity to gefitinib. The EGFRvIII mutation that results
`in amino-acid terminal truncation and constitutive
`activation of the receptor has been reported to be
`less sensitive to small molecule TKIs than the wild-
`type receptor. Moreover, mutations in threonine
`766 in the EGFR kinase domain confer resistance to
`quinazoline inhibitors 22. This site appears to be a
`“hot-spot” for the development of resistance since
`mutation of the corresponding threonines in C-Abl,
`p38 and Src also confer resistance to their respec-
`tive small molecule inhibitors 22. Conversely, data
`also indicate that pre-treatment EGFR mutations can
`increase sensitivity to EGFR blockade by gefitinib 23.
`Preliminary studies indicate that tumour samples
`from 9 NSCLC patients who responded to gefinitib
`were found to contain recurrent heterozygous single
`amino-acid mutations in the EGFR kinase domain (at
`exons 18, 19 and 21) in contrast to samples from
`the patients who did not respond that did not have
`any of these mutations. Mutated EGFRs were more
`sensitive to ligand stimulation and consequently,
`more sensitive to gefinitib.
`Inhibition of EGFR phosphorylation can also be
`uncoupled from inhibition of downstream signalling
`by alterations in other signalling effectors, such as
`constitutively activating mutations of Ras, B-Raf and
`PI3 kinase or loss of expression of the Akt regulator
`PTEN 24 or activation of other tyrosine kinase or G-
`protein coupled receptors (eg other erbB receptors
`or the IGF-IR). Combinations of targeted inhibitors
`need further evaluation in the clinic.
`
`IMATINIB RESISTANCE
`
`Imatinib (GlivecTM, GliveecTM), a small molecule
`inhibitor of BCR-ABL, C-kit and PDGFR-α, has had
`a major impact on the treatment of chronic myeloid
`leukaemia (CML) and gastrointestinal stromal
`
`

`
`10
`
`L. VIDAL - G. ATTARD - S. KAYE - J. DE BONO
`
`tumours (GIST). Several mechanisms of imatinib
`resistance have been reported. Incomplete target
`blockade due to inadequate drug delivery may be a
`reversible resistance mechanism. Reports indicate
`that in chronic phase, Philadelphia chromosome
`positive, CML higher doses of imatinib (600 or 800
`mg daily) may overcome resistance to lower doses
`(400 mg per day) 25. P-glycoprotein (mdr1) overex-
`pression has also been reported to confer resistance
`to imatinib in vitro by reducing intracellular imatinib
`concentrations, although these findings remain con-
`troversial since other reports indicate that imatinib is
`a poor substrate for p-glycoprotein 26,27. Other fac-
`tors that decrease imatinib delivery to its target
`include high levels of alpha-1 acid glycoprotein
`(AGP), which can prevent intracellular penetration
`by binding imatinib 28.
`Probably the most common mechanism of
`acquired imatinib resistance is alteration of the drug
`target 29-32. Gene amplification of BCR-ABL and
`mutations of key amino acids in the ABL or c-Kit
`kinase domains have been reported. Increasing drug
`dose, 25 or concurrently administering Hsp-90
`inhibitors that inhibit the expression of the mature
`target protein, may reverse the impact of BCR-ABL
`gene amplification 32. Approximately 30 different
`point mutations, coding for distinct single amino-acid
`substitutions in the BCR-ABL kinase domain, have
`been isolated from relapsed CML patients resistant
`to imatinib. While it has been suggested that some
`of these mutations predate imatinib therapy, in most
`cases the selective pressure of imatinib treatment
`induces these mutations. Many of these mutations
`have been shown to alter the imatinib-binding region
`of the BCR/ABL kinase domain, thus reducing ima-
`tinib’s binding affinity. These mutations occur either
`at sites that come into direct contact with imatinib
`(T315, F317, and F359), and presumably impair
`drug binding without significantly affecting the bind-
`ing of ATP, or at residues located in distant regions
`implicated in the unique conformational change that
`the kinase domain must undergo to accommodate
`imatinib 29. Crystallographic studies predict that most
`imatinib-resistant mutants should remain sensitive to
`inhibitors that bind ABL with less stringent confor-
`mational requirements than imatinib. Novel Abl
`kinase inhibitors are showing promising activity in
`preclinical studies and are in various stages of devel-
`opment 30. Conversely, however, it is important to
`note that in GIST activating mutations of c-KIT fre-
`quently predate therapy with imatinib and are key
`driving mutations in this disease, predicting response
`and correlating with clinical outcome with imatinib31.
`BCR-ABL signalling may also be inhibited
`through the induction of BCR-ABL protein degrada-
`tion using inhibitors of the molecular chaperone
`Hsp90 or the blockade of critical downstream signal
`transduction proteins 32,33. Hsp90 inhibitors, such as
`17-AAG, have potent BCR-ABL inhibitory activity
`and clinical trials with 17-AAG and imatinib in CML
`
`are underway. Preclinical studies also suggest that
`inhibition of farnesyltransferase, presumably through
`inhibition of Ras downstream signalling, may also
`reverse imatinib resistance 33.
`
`BEVACUZIMAB (AVASTIN™) RESISTANCE
`
`Angiogenesis inhibition has emerged as an
`important therapeutic strategy in the treatment of
`malignancy 34,35. Tumour cells recruit endothelial
`cells by secreting pro-angiogenic factors such as vas-
`cular endothelial growth factor (VEGF) and fibroblast
`growth factor (FGF). VEGF is overexpressed in
`many tumours and is associated with a worse clinical
`outcome. The aetiology of resistance to anti-angio-
`genesis agents has not yet been extensively
`explored. Nonetheless, similar mechanisms of resis-
`tance, as have been described for other targeted
`agents, are likely. Elevated pro-angiogenic ligand (eg
`VEGFs, FGFs) expression may contribute to antian-
`giogenesis drug resistance, and have already been
`reported to contribute to resistance to chemotherapy
`or endocrine therapy 35,36,37. It is plausible that resis-
`tance to bevacuzimab, a humanised monoclonal
`antibody to VEGF that in combination with
`chemotherapy has resulted in a survival advantage in
`colorectal cancer, may result from increased expres-
`sion of VEGF. This may be reversed by increasing
`bevacuzimab dosing. Rational combinations of
`angiogenesis inhibitors may optimise the antitumour
`effects of these therapeutics.
`
`CONCLUSION: REVERSING RESISTANCE
`TO TARGETED DRUGS
`
`Overall, a number of common resistance mecha-
`nisms have been reported for targeted therapeutics.
`These include 1) inadequate target blockade due to
`sub-optimal drug-delivery; 2) altered target expres-
`sion at the DNA (gene amplification), mRNA or pro-
`tein level; 3) an altered target such as a mutated
`kinase domain; 4) modified target regulating pro-
`teins; 5) signalling by alternative proteins (functional
`redundancy) or different signalling pathways. Several
`strategies for the reversal of resistance to targeted
`anticancer therapeutics can therefore be considered
`and may be widely applicable.
`The first consideration to reversal of resistance
`involves the evaluation of whether maximal target
`blockade has been achieved. Increasing drug dosing
`can reverse resistance, although this may be limited
`by toxicity. Increasing drug delivery by decreasing
`intratumoral interstitial pressure through the inhibi-
`tion of PDGF receptor signaling, may also reverse
`resistance 38. Emerging clinical data suggest that dif-
`ferences in the therapeutic efficacy between mono-
`clonal antibodies and small molecules targeting
`EGFR signaling in colorectal cancer may be due, at
`
`

`
`REVERSING RESISTANCE TO TARGETED THERAPY
`
`11
`
`least in part, to inadequate target blockade 20,39. The
`longer half-life of monoclonal antibodies and their
`irreversible target modulation may be a major advan-
`tage for these agents over the small molecule
`inhibitors. Increasing the frequency of administration
`of the small molecules may help circumvent this and
`result in more complete and sustained target inhibi-
`tion. Moreover, irreversible inhibitors may also
`achieve more profound target inhibition.
`An alternative therapeutic strategy for overcom-
`ing resistance to targeted therapeutics involves the
`inhibition of the post-translational maturation of the
`protein target by modulating the chaperone Hsp-90.
`Hsp90 inhibition modulates the expression of many
`proteins such as HER2, Bcr-Abl, HIF-1-alpha, ER,
`AR, Akt and Raf 40. Hsp90 inhibitors are therefore
`now being evaluated in clinical trials in combination
`with targeted therapies such as imatinib in order to
`reverse resistance to these agents.
`Gene mutations are common causes of acquired
`resistance to targeted therapies, usually changing the
`conformation of the tyrosine kinase-binding site.
`Using molecular and crystallography studies to study
`these alterations, novel compounds that can inhibit
`these mutated targets are being developed. Novel
`Bcr-Abl small molecule inhibitors that can inhibit
`imatinib refractory, mutated, Abl kinases are already
`being evaluated in the clinic 30.
`Finally, the highly selective inhibition of a molec-
`ular target may fail to translate into clinical benefit
`due to the existence of alternative proteins or path-
`ways that may abrogate the induction of cell death
`following target blockade, due to either functional
`redundancy or the complexity of the molecular alter-
`ations inherent to the cancer cell. Combinations of
`targeted therapeutics therefore need to be evaluated
`in the clinic in order to inhibit such escape mecha-
`nisms. Combinations of for example imatinib and
`the m-TOR inhibitor RAD-001 are being evaluated
`in imatinib resistant GIST; erlotinib and trastuzumab
`are being evaluated in combination in HER2 positive
`breast cancer as is the farnesyltransferase inhibitor
`SCH66336 with trastuzumab. The clinical evalua-
`tion of combinations of unapproved, investigational,
`targeted therapies remains, however, a major logisti-
`cal challenge since this frequently involves more
`than one industry partner. This has resulted in an
`increasing interest in less selective agents that can
`hit multiple targets. This strategy has proved suc-
`cessful in the treatment of renal cell carcinoma utilis-
`ing the small molecule inhibitor BAY 43-9006 in
`the treatment of clear cell renal cell carcinoma 41.
`BAY 43-9006 targets VEGFR2, Raf kinase, c-Kit
`and PDGFRβ. A combination-based strategy, utilis-
`ing the EGFR inhibitor erlotinib and the antibody to
`VEGF bevacuzimab, has also shown promising effi-
`cacy (both drugs are being developed by Genentech,
`CA) 42. The biology-based development of combina-
`tions of molecular targeted therapies is likely to be
`key in the future treatment of cancer 43.
`
`REFERENCES
`
`1. Hellerstedt BA, Pienta KJ. The current state of hormo-
`nal therapy for prostate cancer. CA Can J Clin 2002; 3: 154-
`79.
`
`2. O’Donnell A, Judson I, Dowsett M, et al. Hormonal
`impact of the 17alpha-hydroxylase/C(17,20)-lyase inhibitor
`abiraterone acetate (CB7630) in patients with prostate cancer.
`Br J Cancer 2004; 12: 2317-25.
`3. Chen CD, Welsbie DS, Tran C, et al. Molecular deter-
`minants of response to antiandrogen therapy. Nature Med
`2004; 10: 33-9.
`4. Taplin M-E, Bubley GJ, Shuster D, et al. Mutations of
`the androgen-receptor gene in metastatic androgen indepen-
`dent prostate cancer. N Engl J Med 1995; 332:1393-98.
`5. Small EJ, Halabi S, Dawson NA, et al. Antiandrogen
`withdrawal alone or in combination with ketoconazole in
`androgen-independent prostate cancer patients: a phase III
`trial (CALGB 9583). J Clin Oncol 2004; 6:1025-33.
`6. Craft N, Shostak Y, Carey M, Sawyers CL. A mechani-
`sm for hormone-independent prostate cancer through modula-
`tion of androgen receptor signaling by the HER-2/neu tyrosi-
`ne kinase. Nature Med 1999; 3: 264-5.
`7. Dorkin TJ, Robinson MC, Marsh C, Bjartell A, Neal
`DE, Leung HY. FGF8 over-expression in prostate cancer is
`associated with decreased patient survival and persists in
`androgen independent disease. Oncogene 1999; 17: 2755-
`61.
`
`8. Culig Z, Hobisch A, Cronauer MV, et al. Androgen
`receptor activation in prostatic tumor cell lines by insulin-like
`growth factor-I, keratinocyte growth factor, and epidermal
`growth factor. Cancer Res 1994; 20: 5474-8.
`9. Rahman M, Miyamoto H, Chang C. Androgen receptor
`coregulators in prostate cancer: mechanisms and clinical impli-
`cations. Clin Cancer Res 2004; 7: 2208-19.
`10. Clarke R, Leonessa F, Welch JN, Skaar TC. Cellular
`and molecular pharmacology of antiestrogen action resistance.
`Pharmacol Rev 2001; 1: 25-71.
`11. Fuqua SAW, Wiltschke C, Zhang QX et al. A
`Hypersensitive Estrogen Receptor- alpha mutation in premali-
`gnant Breast Lesions. Cancer Res 2000; 60: 4026-29.
`12. Nicholson RI, Hutchenson IR, Harper ME, et al.
`Modulation of epidermal growth factor receptor in endocrine-
`resistant, oestrogen receptor-positive breast cancer. Endocr
`Relat Can 2001; 3: 175-82.
`13. Gutteridge E, Gee JM, Nicholson RI, Robertson JFR.
`Biological markers associated with response to gefinitib in
`patients with breast cancer. Proc Am Soc Clin Oncol 2004;
`23: 38.
`14. Robertson JFR, Gutteridge E, Cheung KL, et al.
`Gefitinib is active in acquired tamoxifen (TAM)-resistant
`oestrogen receptor (ER)-positive and ER-negative breast can-
`cer: Results from a phase II study. Proc Am Soc Clin Oncol
`2003; 22: 7.
`15. Johnston SR, Hickish T, Ellis P, et al. Phase II study of
`the efficacy and tolerability of two dosing regimens of the far-
`nesyl transferase inhibitor, R115777, in advanced breast can-
`cer. J Clin Oncol 2003; 13: 2492-9.
`16. Baselga J, Fumaleu P, Gil M, et al. Phase II, 3-arm
`study of CCI-779 in combination with letrozol in post

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