throbber
V O L U M E 2 8 䡠 N U M B E R 5 䡠 F E B R U A R Y 1 0 2 0 1 0
`
`JOURNAL OF CLINICAL ONCOLOGY
`
`O R I G I N A L R E P O R T
`
`From the Center for Sarcoma and Bone
`Oncology and Ludwig Center for
`Cancer Research; and Department of
`Radiology, Dana-Farber Cancer Insti-
`tute; Harvard Medical School; Division
`of Translational Medicine, Departments
`of Medicine and of Pathology, Brigham
`and Women’s Hospital; and Center for
`Molecular Oncologic Pathology, Dana-
`Farber Cancer Institute/Brigham and
`Women’s Cancer Center, Boston, MA;
`and Department of Pathology and
`Melanoma and Sarcoma Service,
`Department of Medicine, Memorial
`Sloan-Kettering Cancer Center, New
`York, NY.
`
`Submitted July 24, 2009; accepted
`September 25, 2009; published online
`ahead of print at www.jco.org on
`January 4, 2010.
`
`Supported in part by the sPECial Fund
`for PEComa Research and by the
`Virginia and D.K. Ludwig Fund for
`Cancer Research (to the Dana-Farber/
`Harvard Ludwig Center).
`
`Presented in part at the 14th Annual
`Meeting of the Connective Tissue
`Oncologic Society,November 13-15,
`2008, London, United Kingdom.
`
`Authors’ disclosures of potential con-
`flicts of interest and author contribu-
`tions are found at the end of this
`article.
`
`Corresponding author: Andrew J.
`Wagner, MD, PhD, Center for Sarcoma
`and Bone Oncology, Dana-Farber
`Cancer Institute, 44 Binney St, Boston,
`MA 02115; e-mail: Andrew_Wagner@
`dfci.harvard.edu.
`
`© 2010 by American Society of Clinical
`Oncology
`
`0732-183X/10/2805-835/$20.00
`
`DOI: 10.1200/JCO.2009.25.2981
`
`Clinical Activity of mTOR Inhibition With Sirolimus in
`Malignant Perivascular Epithelioid Cell Tumors: Targeting
`the Pathogenic Activation of mTORC1 in Tumors
`Andrew J. Wagner, Izabela Malinowska-Kolodziej, Jeffrey A. Morgan, Wei Qin, Christopher D.M. Fletcher,
`Natalie Vena, Azra H. Ligon, Cristina R. Antonescu, Nikhil H. Ramaiya, George D. Demetri,
`David J. Kwiatkowski, and Robert G. Maki
`
`A
`
`B
`
`S
`
`T
`
`R
`
`A
`
`C
`
`T
`
`Purpose
`Perivascular epithelioid cell tumors (PEComas) represent a family of mesenchymal neoplasms,
`mechanistically linked through activation of the mTOR signaling pathway. There is no known
`effective therapy for PEComa, and the molecular pathophysiology of aberrant mTOR signaling
`provided us with a scientific rationale to target this pathway therapeutically. On this mechanistic
`basis, we treated three consecutive patients with metastatic PEComa with an oral mTOR
`inhibitor, sirolimus.
`
`Patients and Methods
`Patients with advanced PEComa were treated with sirolimus and consented to retrospective
`collection of data from their medical records and analysis of archival tumor specimens. Tumor
`response was determined by computed tomography scans obtained at the clinical discretion of the
`treating physicians. Tumors were assessed for immunohistochemical evidence of mTORC1
`activation and genetic evidence of alterations in TSC1 and TSC2.
`
`Results
`Radiographic responses to sirolimus were observed in all patients. PEComas demonstrated loss
`of TSC2 protein expression and evidence of baseline mTORC1 activation. Homozygous loss of
`TSC1 was identified in one PEComa.
`
`Conclusion
`Inhibition of mTORC1, pathologically activated by loss of the TSC1/TSC2 tumor suppressor
`complex, is a rational mechanistic target for therapy in PEComas. The clinical activity of sirolimus
`in PEComa additionally strengthens the pathobiologic similarities linking PEComas to other
`neoplasms related to the tuberous sclerosis complex.
`
`J Clin Oncol 28:835-840. © 2010 by American Society of Clinical Oncology
`
`INTRODUCTION
`
`Sarcomas are a heterogeneous collection of tu-
`mors sharing a common mesenchymal origin. His-
`torically, treatment studies of pooled subtypes of
`sarcoma have shown overall poor responses to con-
`ventional chemotherapeutic agents.1 However, in
`recent years, identification of molecular subtypes of
`sarcoma has led to the application of effective tar-
`geted therapies, such as imatinib mesylate for the
`treatment of gastrointestinal stromal tumors that
`usually harbor activating mutations in the KIT re-
`ceptor tyrosine kinase. Identification of molecular
`alterations in other sarcoma subtypes may lead to
`more effective therapies for this otherwise difficult-
`to-treat group of diseases.
`
`tumor
`The perivascular epithelioid cell
`(PEComa) family of tumors consists of related mes-
`enchymal neoplasms that exhibit myomelanocytic
`differentiation and share a distinctive cell type,
`the perivascular epithelioid cell, or PEC.2-4 The ma-
`jor members of this family include lymphangio-
`leiomyomatosis (LAM), a disease predominantly
`presenting as numerous nodular and interstitial pul-
`monary lesions in premenopausal women; angio-
`myolipoma (AML), commonly identified as an
`asymptomatic renal lesion with evidence of vas-
`cular, muscle, and adipocytic differentiation; and
`PEComa, an epithelioid malignancy with clear-to-
`granular eosinophilic cytoplasm typically arising in
`the gastrointestinal tract, retroperitoneum, uterus,
`or somatic soft tissues, composed of nests and sheets
`
`© 2010 by American Society of Clinical Oncology
`Information downloaded from jco.ascopubs.org and provided by at Univ of Chicago Library on February 25, 2016 from
`
`205.208.61.64Copyright © 2010 American Society of Clinical Oncology. All rights reserved.
`
`835
`
`Ex. 1100-0001
`
`

`
`Wagner et al
`
`of epithelioid or occasionally spindled cells, intimately related to blood
`vessel walls.
`Tumors of the PEComa family are rare and usually occur
`sporadically. LAM and AML also are seen at high frequency in
`patients with tuberous sclerosis complex (TSC), a disorder caused
`by mutation of TSC1 or TSC2, for which the gene products nega-
`tively regulate mTORC1 through inhibition of the mTOR kinase
`activator RHEB. Recently Bissler et al5 described modest, transient
`improvement in lung function and reduction in size of AML in a
`trial of 25 patients with LAM and AML treated with sirolimus, an
`inhibitor of mTORC1. PEComas have rarely been reported in
`association with TSC.6
`Most PEComas are benign tumors and do not recur after com-
`plete surgical resection. However, a subset of PEComas exhibit malig-
`nant behavior, with either locally invasive recurrences or development
`of distant metastases, most commonly in the lung. No effective ther-
`apy for malignant PEComa has been described. Because PEComas
`share activation of the mTOR pathway with LAM and AML in many
`instances,6,7 we treated three consecutive patients at two institutions
`with sirolimus.
`
`PATIENTS AND METHODS
`
`Patient Selection, Treatment, and Clinical Assessments
`Three consecutive patients with PEComa who presented to Dana-
`Farber Cancer Institute or Memorial Sloan-Kettering Cancer Center were
`offered off-label treatment with sirolimus by their treating physicians.
`None of the patients exhibited clinical signs or a history of TSC. There was
`no significant family history of other cancers in any of the index patients.
`All patients provided informed consent for treatment with sirolimus as
`well as retrospective review of medical records and evaluation of archival
`tumor specimens according to institutional review board–approved pro-
`tocols. The dosage of sirolimus (range, 1 mg every other day to 8 mg daily)
`was determined by the treating physician and was adjusted on the basis of
`trough sirolimus levels or patient tolerance. Disease status was assessed by
`CT scans at baseline and at intervals recommended by the treating physi-
`cian. [18F]fluorodeoxyglucose–positron emission tomography scans were
`not performed as part of tumor assessment.
`
`Histologic and Immunohistochemical Evaluations
`Hemotoxylin and eosin staining and immunohistochemical stains of
`sections of archival formalin-fixed paraffin-embedded tissue were per-
`formed according to standard techniques for routine clinical pathologic
`evaluation. For additional immunohistochemical analyses, slides were
`processed through a xylene and ethanol series, subjected to Dako Antigen
`Retrieval procedure (Dako, Carpinteria, CA), and stained with a monoclo-
`nal mouse antiphospho-S6 antibody (S240/244, clone DAK-S6-240; Dako,
`Carpinteria, CA) or a polyclonal rabbit anti-TSC2 antibody (gift of Vijaya
`Ramesh, Massachusetts General Hospital, Boston, MA) followed by horse-
`radish peroxidase detection.
`
`Genetic Analyses
`Genomic DNA was extracted from paraffin-embedded tumor tissue or
`peripheral-blood mononuclear cells and was assessed for copy number
`changes by mulitplex ligation-dependent probe amplification (MLPA) with
`nine exonic probes for TSC1 and TSC2, respectively, and five control probes
`from other genomic locations.8 Signals were normalized to values obtained
`with DNA extracted from control tissues. A value of 1 represented two copies
`of the assayed gene, 0.5 represented one copy, and 0 represented loss of
`both copies.
`
`Fluorescence in situ hybridization (FISH) was performed by using
`interphase nuclei isolated from archival paraffin-embedded tissue and
`fosmid DNA probes corresponding to TSC1 (G248P87270H4 and
`G248P87503E5) and TSC2 (G248P89119C12 and G248P84443G9) with
`4⬘,6-diamidino-2-phenylindole counterstaining, according to standard
`methods.9 Genomic DNA from PEComas was amplified for PIK3CA (ex-
`ons 10 and 21), AKT1 (exon 3), and KRAS (exons 2 and 3), and then was
`sequenced by standard dideoxy sequencing.
`
`A
`
`B
`
`C
`
`Fig 1. Computed tomography images of perivascular epithelioid cell tumors
`(PEComas;
`left panels) at baseline and (right panels) after treatment with
`sirolimus. All
`images are chosen to show the maximal size of the lesions
`indicated by arrows; other lesions may be out of phase in the two studies. (A)
`Retroperitoneal PEComa before and after 1 year of treatment. (B) Renal PEComa
`before and after 9 months of treatment. (C, top) Metastatic uterine PEComa in
`lung before and after 6 weeks of treatment; some lesions are improved, whereas
`others remain stable. (C, bottom, left) Before and (right) 3 weeks after treatment
`with sorafenib.
`
`836
`
`JOURNAL OF CLINICAL ONCOLOGY
`© 2010 by American Society of Clinical Oncology
`Information downloaded from jco.ascopubs.org and provided by at Univ of Chicago Library on February 25, 2016 from
`
`205.208.61.64Copyright © 2010 American Society of Clinical Oncology. All rights reserved.
`
`Ex. 1100-0002
`
`

`
`Sirolimus for Malignant PEComa
`
`RESULTS
`
`Patient outcomes with systemic therapy are summarized in narrative
`format in this Results section. Appendix Table A1 indicates the out-
`comes of systemic therapy for index patients as well.
`
`Patient 1
`Patient 1 is a 65-year-old man who underwent resection of a
`20-cm retroperitoneal PEComa in 2005, complicated by preoper-
`ative tumor rupture. Multifocal retroperitoneal recurrences were
`noted on surveillance imaging, and all sites of disease were resected
`in 2007. Additional sites of disease were noted on surveillance scans
`3 months later.
`He participated in a phase I study of an oral inhibitor of MET and
`developed rapid disease progression. In 2008, he began treatment with
`sirolimus 8 mg daily and achieved a serum trough level of 36 ng/mL;
`sirolimus has been well tolerated except for mild fatigue. Significant
`reduction in size of all tumors was noted on serial CT scans (Fig 1A),
`with near complete disappearance at 1 year. Both treatment and re-
`sponse are ongoing currently at 16 months of follow-up.
`
`Patient 2
`Patient 2 is a 70-year-old man who presented with hematuria
`in 2001. A radical nephrectomy was performed for removal of a
`9-cm renal mass, and the pathology was initially interpreted as a
`poorly differentiated sarcomatoid variant of clear cell carcinoma.
`Five years later, a local recurrence was detected and resected. One
`year later, the patient developed flank pain and again was noted to
`have developed a locoregional recurrence. Treatment with sunitinib
`
`was initiated but was stopped with evidence of disease progression
`after 6 weeks.
`In 2008, the patient presented to one of our institutions, and the
`pathologic diagnosis was reclassified as PEComa. Sirolimus 4 mg daily
`was initiated but was complicated by diarrhea and fatigue. The dose
`was reduced to 1 mg every other day, and improvement in the adverse
`effect profile and reduction in flank pain were noted. Restaging studies
`demonstrated reduction in size of the tumor (Fig 1B), with 40%
`reduction in the longest diameter and continued disease control for 10
`months before evidence of progression despite a sirolimus level of 9.4
`ng/mL. He subsequently had an additional surgical resection and
`remains alive with disease.
`
`Patient 3
`Patient 3 was a 61-year-old woman who presented with uter-
`ine bleeding in 2007. A total abdominal hysterectomy was per-
`formed, which identified a 9-cm PEComa with malignant features
`arising from the cervix. Staging studies demonstrated numerous
`bilateral pulmonary metastases. Sirolimus 4 mg orally each day was
`initiated. Restaging studies performed after 6 weeks of treatment
`revealed interval reduction in size and central cavitation of most of
`the pulmonary nodules (Fig 1C, top). Repeat evaluation at 3
`months demonstrated significant progression of disease; a serum
`trough sirolimus level was 5 ng/mL. The dose of sirolimus was
`increased to 8 mg daily, but restaging studies 1 month later showed
`additional progression of disease, and the serum trough sirolimus
`level was 7 ng/mL. The sirolimus dose was decreased to 2 mg daily,
`and clarithromycin 500 mg daily was added to inhibit CYP3A4, the
`major enzyme responsible for metabolism of sirolimus. The serum
`
`Retroperitoneal
`
`Renal
`
`Uterine
`
`A
`
`B
`
`C
`
`Fig 2. Perivascular epithelioid cell
`tumor (PEComa) histology and immuno-
`histochemical stains. Formalin-fixed paraffin-
`embedded sections stained with (A)
`hematoxylin and eosin, (B) antiphospho-S6
`antibody, and (C) anti-TSC2 antibody.
`
`www.jco.org
`
`© 2010 by American Society of Clinical Oncology
`Information downloaded from jco.ascopubs.org and provided by at Univ of Chicago Library on February 25, 2016 from
`
`205.208.61.64Copyright © 2010 American Society of Clinical Oncology. All rights reserved.
`
`837
`
`Ex. 1100-0003
`
`

`
`Wagner et al
`
`3’
`
`
`
`E22 C5-chr4C4-chr2C3-chr11C2-chr17C1-chr22
`
`
`
`
`Control Probes
`
`5’
`
`E1
`
`E6
`
`E12
`
`E16
`
`E23
`
`E27
`
`E34
`
`E39
`
`TSC2
`
`1.0
`
`0.5
`
`0
`
`1.0
`
`0.5
`
`0
`
`1.0
`
`0.5
`
`0
`
`1.0
`
`0.5
`
`0
`
`A
`
`Assessment Value
`Genomic Copy No.
`
`B
`
`Assessment Value
`Genomic Copy No.
`
`C
`
`Assessment Value
`Genomic Copy No.
`
`D
`
`Assessment Value
`Genomic Copy No.
`
`E
`
`5’
`
`E1
`
`E3
`
`E8
`
`E9
`
`E14
`
`E19
`
`TSC1
`
`Fig 3. Mulitplex ligation-dependent probe amplification (MLPA) and fluorescent
`in situ hybridization (FISH) assays of TSC1 and TSC2 in perivascular epithelioid
`cell tumor (PEComa) samples. Genomic DNA from (A-C) tumor or (D) peripheral
`blood assessed for copy number by mulitplex ligation-dependent probe amplifi-
`cation (MLPA). Signals were normalized to values from control tissues. (E) FISH
`using (green) TSC1-specific DNA probes, (red) chromosome 16 centromeric
`probes, and nuclei isolated from uterine PEComa tissue.
`
`trough sirolimus level increased to 20 ng/mL, and CT scans 1
`month later demonstrated stabilization of the majority of lesions
`and interval reduction in the size of some nodules. Additional
`progression of disease was noted 1 month later, and treatment was
`empirically changed to sorafenib 200 mg twice daily and sirolimus
`4 mg daily. Significant cavitation of lung masses developed within
`2 weeks (Fig 1C, bottom) with improvement in symptoms and
`obviation of the need for supplemental oxygen, although bilateral
`pneumothoraces also developed. After 2 additional months of
`disease control, symptomatic worsening of pulmonary parenchy-
`mal disease ensued, and the patient died.
`
`Correlative Studies
`Hematoxylin and eosin–stained tumor sections showed sheets
`of epithelioid cells with granular eosinophilic cytoplasm, inti-
`mately associated with blood vessels, and with frequent mitoses,
`pleomorphism, and necrosis suggestive of malignant behavior (Fig
`2A). Immunohistochemical stains were positive for melanocytic
`markers HMB45 and Melan-A, variably positive for smooth mus-
`cle actin and desmin, and negative for cytokeratins and S100 (not
`shown). Specimens from all three patients demonstrated strong,
`diffuse, cytoplasmic staining for phosphorylated S6 protein (Fig
`2B) consistent with activation of mTORC1. Lesional cells showed
`loss of expression of tuberin, the gene product of TSC2, whereas
`expression generally was maintained in normal vessel wall vascular
`smooth muscle cells (Fig 2C). The specificity of the anti-TSC2
`antibody was confirmed in TSC2-null and -expressing cell lines
`(Appendix Fig A1).
`The integrity of the TSC1 and TSC2 genomic loci was examined
`by MLPA8 (Fig 3). Genomic DNA was extracted from paraffin-
`embedded tumor tissue (Figs 3A, 3B, and 3C) or peripheral blood (Fig
`3D) and was assessed for copy number changes by MLPA by using
`nine exonic probes for TSC1 and TSC2, respectively, and five control
`probes from other genomic locations.
`This technique allowed genomic copy number assessment; a
`normalized value of 1 indicated two intact alleles, and values of 0.5 and
`0 indicated a single allele and complete genomic loss, respectively.
`Marked reduction in signal from the TSC1 gene was observed in the
`uterine tumor specimen from patient 3 (Figs 3C to D), consistent with
`somatic biallelic deletion of TSC1 (Fig 3A). MLPA demonstrated no
`alterations in tumor samples from the retroperitoneal PEComa (pa-
`tient 1; Fig 3A) or the renal PEComa (patient 2; Fig 3B), which suggests
`that mechanisms other than gross alteration of the TSC1 or TSC2
`genomic loci were important in loss of TSC2 expression in those
`two tumors.
`FISH, performed by using interphase nuclei isolated from
`archival paraffin-embedded tissue and probes corresponding to
`TSC1 and TSC2, confirmed homozygous loss of TSC1 (Fig 3E, green
`signal) in greater than 60% of nuclei from the uterine PEComa sam-
`ple, whereas the centromeric region of chromosome 9 (Fig 3E, red
`signal) was maintained.
`Karyotype analysis of the retroperitoneal PEComa (patient 2),
`previously performed for clinical purposes, showed two copies of
`chromosome 16 as well as an add([16p]; not shown). The add(16p13)
`involved chromosome breakage within band p13, loss of material
`distal to the break, and addition of unidentified chromosomal mate-
`rial in its place. Because TSC2 maps to 16p13, interphase FISH was
`
`838
`
`JOURNAL OF CLINICAL ONCOLOGY
`© 2010 by American Society of Clinical Oncology
`Information downloaded from jco.ascopubs.org and provided by at Univ of Chicago Library on February 25, 2016 from
`
`205.208.61.64Copyright © 2010 American Society of Clinical Oncology. All rights reserved.
`
`Ex. 1100-0004
`
`

`
`Sirolimus for Malignant PEComa
`
`performed to determine the TSC2 copy number. Three copies of TSC2
`were detected (not shown), which suggests that the breakpoint of the
`add(16) maps distal to the TSC2 probe. The possibility of small dele-
`tions of or point mutations in TSC2, which are undetectable by FISH,
`cannot be excluded. No other evidence of deletion or rearrangement
`of either TSC1 or TSC2 was observed.
`PEComa DNA samples were sequenced to search for known
`activating mutations in the PIK3CA and AKT1 genes as well as for the
`common mutations that occur in KRAS. No mutations were detected
`in any of the three PEComas in any of these three genes.
`
`DISCUSSION
`
`Metastatic PEComa is a rare form of sarcoma for which no effective
`therapy has been described previously and which has a uniformly fatal
`outcome. We have observed significant clinical responses in three
`patients treated with sirolimus, an inhibitor of mTORC1, including
`one patient with an ongoing near complete response of greater than 14
`months duration. Tumors from all three patients showed evidence of
`mTORC1 activation, and somatic deletion of TSC1 was identified as
`the likely mechanism in patient 3. TSC1 acts to stabilize TSC2, so its
`genomic loss will lead to lack of TSC2 expression.10 The mechanisms
`of mTOR activation in patients 1 and 2 are unclear, but they poten-
`tially include small deletions or inactivating or missense mutations in
`TSC1 or TSC2 that would similarly account for loss of TSC2 expres-
`sion in those tumors.
`Other evidence to support activation of the mTOR pathway in
`PEComas also has recently been described. Kenerson et al7 reported
`immunohistochemical evidence of mTORC1 activity in 15 PEComas
`and absence of AKT phosphorylation in 14 tumors, which suggests the
`loss of TSC1 or TSC2 as potential mechanisms. Similarly, Pan et al6
`described elevated phospho-p70S6K and reduced phospho-AKT in 11
`of 12 PEComas.6 Seven of these tumors had loss of heterozygosity of
`the TSC2 region, and one additionally showed loss of heterozygosity
`of TSC1.
`Our data are consistent with findings to date of the activity of
`mTOR inhibitors in tumors known to be biologically related to
`PEComas, specifically AML and LAM. After case reports of pa-
`tients with renal AML and LAM responding to sirolimus were
`published,11-13 Bissler et al5 reported on treatment of 25 patients with
`AML or LAM with sirolimus for 12 months followed by 12 months of
`observation.5 After 12 months of therapy, AML volume decreased
`53% but returned to 86% of baseline after the year of observation,
`which indicated the need for continued inhibition to maintain
`tumor shrinkage. Less-impressive improvements in respiratory
`function were observed in patients with LAM, which also reversed
`on observation alone. Interestingly, facial angiofibromas associ-
`ated with tuberous sclerosis also have significantly improved on
`sirolimus in a case report.14 There are presently no reports of
`mTOR inhibitors as treatment of clear cell “sugar” tumors of the
`pancreas or lung, another neoplasm considered to be a PEComa,
`but it is expected that such tumors would respond to mTOR-
`directed therapy as well.
`The efficacy of mTOR inhibitors has been explored in patients
`with a heterogenous mix of other metastatic sarcomas, in each case
`with only a modest response rate.15,16 However, the status of
`mTOR activation of these sarcomas is unknown, although in one
`
`study the presence of S6 phosphorylation correlated with a higher
`likelihood of disease control with an mTOR inhibitor.17 Taken to-
`gether, these observations suggest that activation of mTOR through
`loss of the TSC1/TSC2 repressor complex, or potentially by other
`means, is likely a common and critically pathogenic event in PEComas.
`Inhibition of mTOR has resulted in significant clinical activity
`in patients with PEComa and merits additional investigation in a
`prospective study. Absence of immunohistochemical evidence of
`TSC2 expression or the less-specific presence of S6 phosphoryla-
`tion may be predictive markers for responsiveness to inhibitors
`of mTORC1. These findings additionally unify the concept of
`PEComa, AML, and LAM as closely related pathologic entities,
`from histology to genetic changes to evidence of therapeutic ben-
`efit from mTOR blockade.
`
`AUTHORS’ DISCLOSURES OF POTENTIAL CONFLICTS
`OF INTEREST
`
`Although all authors completed the disclosure declaration, the following
`author(s) indicated a financial or other interest that is relevant to the subject
`matter under consideration in this article. Certain relationships marked
`with a “U” are those for which no compensation was received; those
`relationships marked with a “C” were compensated. For a detailed
`description of the disclosure categories, or for more information about
`ASCO’s conflict of interest policy, please refer to the Author Disclosure
`Declaration and the Disclosures of Potential Conflicts of Interest section in
`Information for Contributors.
`Employment or Leadership Position: None Consultant or Advisory
`Role: Andrew J. Wagner, Genentech (C); George D. Demetri, Ariad (C),
`Novartis (C), Pfizer (C), Genentech (C); Robert G. Maki, Novartis (C)
`Stock Ownership: None Honoraria: Robert G. Maki, Novartis Research
`Funding: Andrew J. Wagner, Genentech; Robert G. Maki, Novartis,
`Pfizer Expert Testimony: George D. Demetri, Ariad (U) Other
`Remuneration: None
`
`AUTHOR CONTRIBUTIONS
`
`Conception and design: Andrew J. Wagner, Jeffrey A. Morgan,
`Christopher D.M. Fletcher, Azra H. Ligon, George D. Demetri, David J.
`Kwiatkowski, Robert G. Maki
`Financial support: Andrew J. Wagner, George D. Demetri
`Administrative support: Andrew J. Wagner, George D. Demetri
`Provision of study materials or patients: Andrew J. Wagner, Jeffrey A.
`Morgan, Christopher D.M. Fletcher, Cristina R. Antonescu,
`Robert G. Maki
`Collection and assembly of data: Andrew J. Wagner, Izabela
`Malinowska-Kolodziej, Jeffrey A. Morgan, Wei Qin, Christopher D.M.
`Fletcher, Natalie Vena, Azra H. Ligon, Cristina R. Antonescu, Nikhil H.
`Ramaiya, David J. Kwiatkowski, Robert G. Maki
`Data analysis and interpretation: Andrew J. Wagner, Izabela
`Malinowska-Kolodziej, Jeffrey A. Morgan, Christopher D.M. Fletcher,
`Natalie Vena, Azra H. Ligon, Nikhil H. Ramaiya, David J. Kwiatkowski,
`Robert G. Maki
`Manuscript writing: Andrew J. Wagner, Izabela Malinowska-Kolodziej,
`Christopher D.M. Fletcher, Azra H. Ligon, George D. Demetri, David J.
`Kwiatkowski, Robert G. Maki
`Final approval of manuscript: Andrew J. Wagner, Izabela
`Malinowska-Kolodziej, Jeffrey A. Morgan, Wei Qin, Christopher D.M.
`Fletcher, Natalie Vena, Azra H. Ligon, Cristina R. Antonescu, Nikhil H.
`Ramaiya, George D. Demetri, David J. Kwiatkowski, Robert G. Maki
`
`www.jco.org
`
`© 2010 by American Society of Clinical Oncology
`Information downloaded from jco.ascopubs.org and provided by at Univ of Chicago Library on February 25, 2016 from
`
`205.208.61.64Copyright © 2010 American Society of Clinical Oncology. All rights reserved.
`
`839
`
`Ex. 1100-0005
`
`

`
`REFERENCES
`
`1. Wagner A: Treatment of advanced soft tis-
`sue sarcoma: Conventional agents and promising
`new drugs. J Natl Compr Canc Netw 5:401-410,
`2007
`2. Hornick JL, Fletcher CD: PEComa: What do
`we know so far? Histopathology 48:75-82, 2006
`3. Folpe AL, Kwiatkowski DJ: Perivascular epi-
`thelioid cell neoplasms: Pathology and pathogenesis.
`Hum Pathol 10.1016/j.humpath.2009.05.011 [epub
`ahead of print on July 15, 2009]
`4. Folpe AL, Mentzel T, Lehr HA, et al: Perivas-
`cular epithelioid cell neoplasms of soft tissue and
`gynecologic origin: A clinicopathologic study of 26
`cases and review of the literature. Am J Surg Pathol
`29:1558-1575, 2005
`5. Bissler JJ, McCormack FX, Young LR, et al:
`Sirolimus for angiomyolipoma in tuberous sclerosis
`complex or lymphangioleiomyomatosis. N Engl J Med
`358:140-151, 2008
`6. Pan CC, Chung MY, Ng KF, et al: Constant
`allelic alteration on chromosome 16p (TSC2 gene)
`
`Wagner et al
`
`in perivascular epithelioid cell tumour (PEComa):
`Genetic evidence for the relationship of PEComa
`with angiomyolipoma. J Pathol 214:387-393, 2008
`7. Kenerson H, Folpe AL, Takayama TK, et al:
`Activation of the mTOR pathway in sporadic angio-
`myolipomas and other perivascular epithelioid cell
`neoplasms. Hum Pathol 38:1361-1371, 2007
`8. Kozlowski P, Roberts P, Dabora S, et al: Identi-
`fication of 54 large deletions/duplications in TSC1 and
`TSC2 using MLPA, and genotype-phenotype correla-
`tions. Hum Genet 121:389-400, 2007
`9. Firestein R, Bass AJ, Kim SY, et al: CDK8 is a
`colorectal cancer oncogene that regulates beta-
`catenin activity. Nature 455:547-551, 2008
`10. Benvenuto G, Li S, Brown SJ, et al: The
`tuberous sclerosis-1 (TSC1) gene product hamartin
`suppresses cell growth and augments the expres-
`sion of the TSC2 product tuberin by inhibiting its
`ubiquitination. Oncogene 19:6306-6316, 2000
`11. Herry I, Neukirch C, Debray MP, et al: Dra-
`matic effect of sirolimus on renal angiomyolipomas
`in a patient with tuberous sclerosis complex. Eur
`J Intern Med 18:76-77, 2007
`
`■ ■ ■
`
`12. Taille´ C, Debray MP, Crestani B: Sirolimus
`treatment for pulmonary lymphangioleiomyomato-
`sis. Ann Intern Med 146:687-688, 2007
`13. Wienecke R, Fackler I, Linsenmaier U, et al:
`Antitumoral activity of rapamycin in renal angiomyo-
`lipoma associated with tuberous sclerosis complex.
`Am J Kidney Dis 48:e27-e29, 2006
`14. Hofbauer GF, Marcollo-Pini A, Corsenca A, et al:
`The mTOR inhibitor rapamycin significantly improves
`facial angiofibroma lesions in a patient with tuberous
`sclerosis. Br J Dermatol 159:473-475, 2008
`15. Chawla SP, Tolcher AW, Staddon AP, et al:
`Updated results of a phase II trial of AP23573, a
`novel mTOR inhibitor, in patients with advanced soft
`tissue or bone sarcomas. J Clin Oncol 24:521s, 2006
`(suppl; abstr 9505)
`16. Okuno SH, Mahoney MR, Bailey HH, et al: A
`multicenter phase II consortium study of the mTOR
`inhibitor CCI-779 in advanced soft tissue sarcomas.
`J Clin Oncol 24:521s, 2006 (suppl; abstr 9504)
`17. Iwenofu OH, Lackman RD, Staddon AP, et al:
`Phospho-S6 ribosomal protein: A potential new pre-
`dictive sarcoma marker for targeted mTOR therapy.
`Mod Pathol 21:231-237, 2008
`
`Be the First to Hear When New Clinical Cancer Research is Published Online
`By signing up for JCO’s Early Release Notification, you will be alerted and have access to new articles posted online
`every Monday, weeks before they appear in print. All Early Release articles are searchable and citable, and are posted
`on JCO.org in advance of print publication. Simply go to jco.org/earlyrelease, sign in, select “Early Release Notification,”
`and click the SUBMIT button. Stay informed—sign up today!
`
`840
`
`JOURNAL OF CLINICAL ONCOLOGY
`© 2010 by American Society of Clinical Oncology
`Information downloaded from jco.ascopubs.org and provided by at Univ of Chicago Library on February 25, 2016 from
`
`205.208.61.64Copyright © 2010 American Society of Clinical Oncology. All rights reserved.
`
`Ex. 1100-0006

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