throbber
Am J Physiol Renal Physiol 306: F279–F283, 2014.
`First published November 13, 2013; doi:10.1152/ajprenal.00525.2013.
`
`Review
`
`Tuberous sclerosis complex, mTOR, and the kidney: report of an
`NIDDK-sponsored workshop
`
`Elizabeth P. Henske,1 Rebekah Rasooly,2 Brian Siroky,3 and John Bissler4
`1Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston,
`Massachusetts; 2Division of Kidney, Urologic, and Hematologic Diseases, National Institute of Diabetes and Digestive and
`Kidney Diseases, National Institutes of Health, Bethesda, Maryland; 3Division of Nephrology and Hypertension, Cincinnati
`Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio; and 4Tuberous Sclerosis Complex Center of
`Excellence, Le Bonheur Children’s Hospital, University of Tennessee College of Medicine, Memphis Tennessee
`
`Submitted 9 October 2013; accepted in final form 7 November 2013
`
`Downloaded from
`
`http://ajprenal.physiology.org/
`
` by 10.220.33.4 on January 8, 2017
`
`Henske EP, Rasooly R, Siroky B, Bissler J. Tuberous sclerosis complex,
`mTOR, and the kidney: report of an NIDDK-sponsored workshop. Am J Physiol
`Renal Physiol 306: F279 –F283, 2014. First published November 13, 2013;
`doi:10.1152/ajprenal.00525.2013.—Remarkable basic and translational advances
`have elucidated the role of the mammalian target of rapamycin (mTOR) signaling
`network in the pathogenesis of renal disease. Many of these advances originated
`from studies of the genetic disease tuberous sclerosis complex (TSC), leading to
`one of the clearest therapeutic opportunities to target mTOR with rapamycin and its
`analogs (“rapalogs”), which effectively inhibit mTOR complex 1 (mTORC1) by an
`allosteric mechanism. Clinical trials based on these discoveries have provided
`strongly positive therapeutic results in TSC (Bissler JJ, McCormack FX, Young
`LR, Elwing JM, Chuck G, Leonard JM, Schmithorst VJ, Laor T, Brody AS, Bean
`J, Salisbury S, Franz DN. N Engl J Med 358: 140 –151, 2008; Krueger DA, Care
`MM, Holland K, Agricola K, Tudor C, Mangeshkar P, Wilson KA, Byars A,
`Sahmoud T, Franz DN. N Engl J Med 363: 1801–1811, 2010; McCormack FX,
`Inoue Y, Moss J, Singer LG, Strange C, Nakata K, Barker AF, Chapman JT,
`Brantly ML, Stocks JM, Brown KK, Lynch JP 3rd, Goldberg HJ, Young LR,
`Kinder BW, Downey GP, Sullivan EJ, Colby TV, McKay RT, Cohen MM, Korbee
`L, Taveira-DaSilva AM, Lee HS, Krischer JP, Trapnell BC. N Engl J Med 364:
`1595–1606, 2011). In June 2013, the National Institute of Diabetes and Digestive
`and Kidney Diseases convened a small panel of physicians and scientists working
`in the field to identify key unknowns and define possible “next steps” in advancing
`understanding of TSC- and mTOR-dependent renal phenotypes. TSC-associated
`renal disease, which affects ⬎85% of TSC patients, and was a major topic of
`discussion, focused on angiomyolipomas and epithelial cysts. The third major topic
`was the role of mTOR and mTOR inhibition in the pathogenesis and therapy of
`chronic renal disease. Renal cell carcinoma, while recognized as a manifestation of
`TSC that occurs in a small fraction of patients, was not the primary focus of this
`workshop and thus was omitted from panel discussions and from this report.
`
`angiomyolipoma; cyst; mTOR; tuberous sclerosis complex
`
`Angiomyolipomas
`
`Background. ANGIOMYOLIPOMAS can arise sporadically or as
`part of the tuberous sclerosis complex (TSC). Sporadic angio-
`myolipomas can be associated with translocations involving
`the TFE3 transcription factor (1) or the TSC genes (5). The
`panel focused primarily on TSC-associated angiomyolipomas.
`Angiomyolipomas are benign mesenchymal tumors composed
`of fat, smooth muscle, and abnormal vascular elements, all of
`which are known to arise from a common precursor cell (13).
`TSC patients typically have multiple, bilateral angiomyolipo-
`mas that can result in renal insufficiency, and these lesions
`
`Address for reprint requests and other correspondence: E. P. Henske, Karp
`Bldg., 6th Floor, One Blackfan Circle, Boston, MA 02115 (e-mail: EHenske
`@Partners.org).
`
`http://www.ajprenal.org
`
`have abnormal vasculature that can form aneurysms that can
`spontaneously hemorrhage, sometimes with life-threatening
`consequences (2). Clinical trials have clearly demonstrated that
`angiomyolipomas shrink in response to inhibitors of mamma-
`lian target of rapamycin complex 1 (mTORC1) (3, 4, 6, 8) and
`that the majority of the tumors return to original volume when
`the treatment is discontinued (4). Thus most patients appear to
`require continuous therapy to suppress angiomyolipoma size.
`Since the rapalog sirolimus typically induces a cytostatic re-
`sponse, it is likely that the decrease in tumor volume does not
`correlate with extensive apoptosis. However, because angiomyo-
`lipomas are highly vascular, mTORC1 inhibition could induce an
`apoptotic cellular response via an antiangiogenic mechanism.
`The allosteric inhibitors (rapalogs) and catalytic inhibitors
`(that impact both mTORC1 and 2) are different, with the
`catalytic inhibitors being much more effective in inhibiting
`
`F279
`
`West-Ward Exhibit 1110
`Henske 2014
`Page 001
`
`

`

`Downloaded from
`
`http://ajprenal.physiology.org/
`
` by 10.220.33.4 on January 8, 2017
`
`Review
`
`F280
`
`TSC, mTOR, AND THE KIDNEY
`
`mTOR activities based on in vitro studies. Preclinical work
`found equivalent benefit of allosteric and catalytic mTOR
`inhibitors in a murine model of TSC (10), but no clinical
`studies of the mTOR catalytic inhibitors involving TSC pa-
`tients have been completed.
`Key unanswered questions. The panel considered key issues
`that remain unaddressed related to the natural history and
`pathogenesis of angiomyolipomas, the evaluation/optimization
`of current angiomyolipoma therapies, and the development of
`novel angiomyolipoma treatment
`strategies, which were
`deemed essential to future progress. These issues are summa-
`rized in Fig. 1.
`Highest priority translational research initiatives. Multiple
`translational approaches were discussed by the panel to address
`the most critical unanswered questions outlined in Fig. 1. The
`highest priority initiatives are the following: 1) genetic analy-
`ses of angiomyolipomas from individual patients, including
`both indolent and rapidly growing tumors and including both
`the solid and vascular components; 2) identification of the
`cell-of-origin of angiomyolipomas, which could facilitate the
`development of animal models of angiomyolipomas; and 3) de-
`velopment of additional cell culture models of angiomyolipomas
`with bi-allelic TSC2 inactivation to facilitate translational and
`preclinical therapeutic advances.
`
`Highest priority clinical research initiatives. In parallel,
`multiple clinical research approaches were discussed to address
`unmet needs in the clinical care of individuals with angiomyo-
`lipomas. From these discussions, the highest priorities are the
`following.
`1) Identification of biomarkers to quantitatively and sensi-
`tively measure angiomyolipoma size and composition. Current
`technology does not easily quantitate the percentage of fat or
`smooth muscle components so that imaging between time
`points or patients can be accurately compared. Both imaging
`and “liquid” (plasma or urinary) biomarkers could be pivotal in
`monitoring angiomyolipoma burden of disease and therapeutic
`response. Imaging modalities that measure the effects of
`mTORC1 activation on cellular metabolism are particularly
`attractive. Serum biomarkers such as VEGF-D, which is a
`diagnostic biomarker and an indicator of therapeutic response
`in lymphangioleiomyomatosis (LAM) (25), a disease that
`shares cellular and genetic features with angiomyolipomas
`(12), should be sought, and markers like collagen IV should be
`pursued (3). Sensitive and specific biomarkers would facilitate
`trials to determine optimal dosing and duration of therapy, and
`to test novel therapies that might result in durable responses,
`ultimately resulting in shorter treatment intervals with associ-
`ated cost savings and a decreased risk of adverse effects.
`
`Natural History and
`Pathogenesis of Angiomyolipomas
`
`Evaluation/Optimization of Current
`Angiomyolipoma Therapies
`
`Development of Novel
`Angiomyolipoma Treatment Strategies
`
`(cid:129) Can early intervention with Rapalogs
` prevent the development of
`
` angiomyolipomas?
`(cid:129) Can addition of other therapeutic agents to
` Rapalogs produce a more robust and
` durable therapeutic response?
`
`(cid:129) Can mTORC1 inhibitors be targeted
`
` directly to angiomyolipomas, thereby
` avoiding systemic toxicity and increasing
` the dose delivered?
`(cid:129) Will catalytic inhibitors of mTOR, which
` inhibit both mTORC1 and mTORC2, be
` more effective than Rapalogs for
`
` angiomyolipoma therapy?
`(cid:129) Does mTOR-dependent feedback to AKT
` and MEK impact the clinical response to
` Rapalogs?
`(cid:129) How does activation of autophagy impact
` the therapeutic response to Rapalogs?
`
`(cid:129) Are angiomyolipomas “locally metastatic,”
` such that a single lesion can seed other
` lesions within the kidney?
`
`(cid:129) Are there genomic or epigenetic
` differences between indolent and locally
` aggressive angiomyolipomas?
`(cid:129) Is there a developmental window during
` which angiomyolipomas initiate?
`(cid:129) What are the mechanisms of aneurysm
` formation in angiomyolipomas?
`
`(cid:129) Are there gender differences in
` angiomyolipoma incidence, size, rate of
` growth, timing of growth, or risk of
`
` bleeding?
`(cid:129) Does blood pressure correlate with the risk
` of hemorrhage from angiomyolipomas?
`(cid:129) Can angiomyolipoma cells be detected in
` the circulation, as has been already shown
` in LAM, providing a “liquid biopsy” allowing
` cellular features of angiomyolipomas to be
` monitored in “real time.”
`
`(cid:129) Why is there such a variable response to
` mTORC1 inhibition?
` Some angiomyolipomas shrink in size
` while others do not, even within the
`
` same kidney.
` Does this reflect the composition of the
` tumor (fat-containing versus fat-poor) or
` its vasculature?
`
`(cid:129) Why do some tumors regrow when
` treatment is discontinued while others
` do not?
` What are the kinetics of the response,
` and is the earliest response vascular or
` cellular?
`(cid:129) What is the mechanism of response?
` Does the decrease in angiomyolipoma size
` with mTORC1 inhibition reflect primarily a
` reduction in individual cell volume, since
` mTORC1 is a key regulator of cell size, or
` a decrease in cell number?
`(cid:129) What is the minimum dose and duration of
` mTOR inhibitor therapy required to induce
` and maintain a reduction in angio-
`
` myolipoma size?
`(cid:129) What are the long-term benefits and risks
` of mTORC1 inhibitor therapy for
`
` angiomyolipomas, including the risk of
` hemorrhage, the impact on the
`
` aneurysmal vessels associated with
`
` angiomyolipomas, and the potential
`
` immunosuppressive effects?
`
`Fig. 1. Unanswered angiomyolipoma issues.
`
`AJP-Renal Physiol • doi:10.1152/ajprenal.00525.2013 • www.ajprenal.org
`
`West-Ward Exhibit 1110
`Henske 2014
`Page 002
`
`

`

`Downloaded from
`
`http://ajprenal.physiology.org/
`
` by 10.220.33.4 on January 8, 2017
`
`TSC, mTOR, AND THE KIDNEY
`
`Review
`
`F281
`
`Biomarkers may also identify the subset of angiomyolipomas
`with a more aggressive clinical phenotype, prioritizing patients
`with these tumors for earlier therapeutic intervention.
`2) There is an additional need for biomarkers that predict the
`risk of hemorrhage, which is a potentially life-threatening
`complication of angiomyolipomas. MRI analysis of aneurysm
`size/complexity may help define imaging biomarkers. Work
`with vascular-related markers may be helpful, and understand-
`ing the biology of aneurysms in the context of TSC will likely
`prove to be a critical step (3, 16, 24).
`3) Genome-wide studies to identify factors that modify the
`risk of angiomyolipoma development, the risk of angiomyoli-
`poma severity, the risk of angiomyolipoma hemorrhage, and/or
`the response to mTORC1 inhibitor therapy should be per-
`formed.
`4) The immunosuppressive effects of mTORC1 inhibitors,
`when used alone in TSC and related diseases, are unclear. The
`risk of infectious complications appears to be low, based on
`available data, but needs to be clarified through longer term
`observational studies.
`5) Natural history studies to identify the earliest develop-
`ment of angiomyolipomas, to determine whether spurts of
`growth can be defined in childhood, puberty, or young adult-
`hood, whether there are gender differences in the timing of
`angiomyolipoma growth, and whether bleeding risk and/or
`growth are correlated with clinical parameters including blood
`pressure, will be pivotal to future prevention trials.
`6) Designing and conducting a placebo-controlled, early-
`intervention study of mTORC1 inhibition for angiomyolipo-
`mas to evaluate its effectiveness as preventative therapy.
`7) Conducting clinical trials to evaluate agents that could
`synergize with mTORC1 inhibitors to produce more effective
`treatment, e.g., Hsp90 inhibitors, autophagy inhibitors, and
`mTOR kinase inhibitors.
`8) Determining the potential benefit of low-dose long-term
`therapies on inhibiting angiomyolipoma development, e.g.,
`
`metformin, NSAIDs, and low-dose or intermittent rapalog
`therapy.
`
`Renal Cystic Disease in TSC
`
`Background. Renal cystic disease in TSC is common, af-
`fecting ⬃50% of patients, ranging in severity from a single
`cyst to multiple, bilateral cystic disease (7, 9, 17). Individuals
`with the contiguous gene syndrome who carry deletions of both
`TSC2 and the adjacent PKD1 gene (⬍5% of TSC patients) can
`even develop severe very early onset polycystic kidney disease
`(18). The mTORC1 pathway has been implicated in the patho-
`genesis of renal cystic disease in autosomal dominant polycys-
`tic kidney disease (ADPKD) (21). To date, the use of mTOR
`inhibitors in treatment of ADPKD has yielded equivocal results
`(4, 5). The timing of mTORC1 inhibition may be pivotal in the
`response of ADPKD-associated cysts to therapy (20, 23).
`Defects in the primary cilium have been observed in TSC-
`deficient cells, including increased ciliary length (11). Together
`with the prominent cystic disease, these findings suggest that
`TSC may be a ciliopathy, yet the role of cilia in the initiation
`of, progression of, and therapy for renal cystic disease in TSC
`is not yet established.
`Key unanswered questions. The panel considered key issues
`involving TSC renal cystic disease that remain unaddressed
`and are essential to future progress. These issues are summa-
`rized in Fig. 2.
`Highest priority translational research initiatives. Multiple
`translational approaches to TSC renal cystic disease were
`discussed by the panel, with the highest priority initiatives
`being the following: 1) comprehensive genetic analysis of a
`large cohort of TSC patients with the polycystic kidney phe-
`notype to define in greater detail the spectrum of mutations that
`cause this manifestation and determine whether mutation of the
`contiguous TSC2 and PKD1 genes account for all of these
`patients; 2) development of an animal model of the TSC2/
`
`(cid:129) The therapeutic response of TSC-associated renal cysts to Rapalogs is largely unknown because the angiomyolipoma trials were not
` designed to assess cystic disease. A particularly important consideration is whether mTORC1 inhibitor therapy can prevent chronic
` kidney disease in patients with the TSC2/PKD1 contiguous gene deletion syndrome.
`(cid:129) The origin, developmental timing and natural history of cyst initiation and progression in TSC are poorly understood. To what extent do
` cysts initiate pre- and post-natally, and what factors and mechanisms are involved in their expansion?
` Do cysts in TSC originate from different segments of the kidney at different developmental time points?
` How does cyst formation and progression relate to angiomyolipoma development?
`(cid:129) The optimal therapeutic endpoints for cystic response are not defined. How should proliferation vs. secretion be considered when
` considering the response of cysts to therapy?
`(cid:129) Factors that may promote cyst progression are not defined. Is hypertension in TSC correlated with cystic disease?
` Does renal injury contribute to cyst progression?
`(cid:129) What accounts for the severity of cystic disease in the TSC2/PKD1 contiguous gene syndrome?
` The impact of co-deletion of TSC2 and PKD1 on a single copy of chromosome 16 in humans seems strikingly different than mutational
` inactivation of each gene on different copies of chromosome 16 in mouse models. Can a mouse model be generated with loss of both
` genes on a single allele to address this?
`(cid:129) There is considerable phenotypic variation in the degree of cyst formation in TSC patients. What is the basis for this variability and is it
` possible to carry out genetic studies of patients at the extremes of the phenotypic distribution?
`(cid:129) What are the functional consequences of ciliary dysfunction in TSC kidneys, and how does this contribute to cystogenesis?
`
`Fig. 2. Unanswered renal cystic disease issues.
`
`AJP-Renal Physiol • doi:10.1152/ajprenal.00525.2013 • www.ajprenal.org
`
`West-Ward Exhibit 1110
`Henske 2014
`Page 003
`
`

`

`Downloaded from
`
`http://ajprenal.physiology.org/
`
` by 10.220.33.4 on January 8, 2017
`
`Review
`
`F282
`
`TSC, mTOR, AND THE KIDNEY
`
`PKD1 contiguous gene deletion syndrome; 3) definition of the
`developmental timing and nephron segment-of-origin of cysts
`in TSC; 4) further examination of the connections between
`ciliary function and cyst formation in TSC; and 5) identifica-
`tion of factors (hypertension, injury) that promote cystogenesis
`in animal models of TSC.
`Highest priority clinical research initiatives. Multiple clin-
`ical research approaches to TSC renal cystic disease were
`considered with the highest priorities being 1) design of clin-
`ical trials to specifically determine whether and how cysts in
`TSC respond to rapalog therapy, including individuals with the
`TSC2/PKD1 contiguous gene syndrome, as the therapeutic
`response of the cystic disease has not been evaluated as an end
`point
`in previous studies; and 2) identification of factors
`including hypertension and renal injury that may promote cyst
`progression in TSC.
`
`Intrinsic Renal Disease Related to mTORC1 Inhibition
`
`Background. Although rapamycin appears to be minimally
`nephrotoxic when used alone, most of the data from humans
`are from studies in which it was used in combination with
`cyclosporine. Rapamycin was not associated with a significant
`increase in proteinuria during the EXIST2 trial of the rapalog
`everolimus for angiomyolipomas (3). However, this trial was
`of relatively short duration and included just over 100 patients.
`Thus the long-term effects of rapalogs as single agents on the
`kidney are not entirely understood. Prolonged treatment with
`mTORC1 inhibitors reduces the total expression of mTOR, as
`well as the expression of rictor and thus mTORC2 formation
`(19). Podocyte expression of nephrin, transient receptor poten-
`tial cation channel 6, and the cytoskeletal adaptor protein Nck
`are significantly decreased following prolonged exposure to an
`mTORC1 inhibitor (22). Furthermore, mTORC1 inhibition
`reduces podocyte adhesion and motility. Together, these ef-
`fects may have a long-term impact on the glomerular and
`tubular structures and deserve attention.
`Key unanswered questions. The panel considered key issues
`involving intrinsic renal disease related to mTORC1 that re-
`main unaddressed and that are essential to be understood as
`treatment may be prolonged. Questions that were discussed
`included the following. Does prolonged rapalog therapy induce
`proteinuria and/or other glomerular or tubular effects in hu-
`mans? Are there differential effects of mTORC1 vs. mTORC2
`inhibition on the kidney that could be relevant to future clinical
`trials involving catalytic mTOR kinase inhibitors?
`Highest priority translational and clinical research initiatives.
`Given that mTORC1 inhibitor therapy will be used in both
`children and adults with TSC and that there are many un-
`knowns related to the long-term impact on the kidney, the
`panel concluded that renal function and proteinuria should be
`monitored in a standardized, prospective manner in individuals
`receiving long-term rapalog therapy.
`
`Conclusions
`
`In summary, there was consensus that areas of high priority
`related to the roles of mTOR in renal disease include the
`following.
`Preclinical models of angiomyolipomas and renal cystic
`disease. Priorities for which the panel had clear consensus
`included identifying the cell-of-origin of angiomyolipomas,
`
`developing mouse models of angiomyolipomas, and develop-
`ing of additional cell lines derived from angiomyolipomas. A
`mouse model that recapitulates the severe, early-onset cystic
`disease observed in the TSC2/PKD1 contiguous gene syn-
`drome is likewise required. Further
`investigation of
`the
`nephron segment-of-origin, the developmental timing of cystic
`disease in TSC, and the role of ciliary dysfunction in TSC-
`associated cystogenesis is critical to the development of tar-
`geted therapy for TSC-associated renal cystic disease. It is also
`important to develop cell culture models for study of TSC
`using induced pluripotent stem cells (iPSC) from patients.
`Biomarkers. The panel determined that future research fo-
`cused on developing and refining imaging techniques to mon-
`itor disease progression, evaluating responses to therapy, and
`providing valuable natural history data would address key
`unmet needs. Focus areas included modalities that would allow
`more precise monitoring of tumor size, imaging with novel
`PET tracers that would allow differentiation between fat and
`other elements within angiomyolipomas, and serum biomark-
`ers of disease burden and therapeutic response are crucial.
`Similarly,
`imaging and biochemical biomarkers that could
`prognosticate and monitor therapy for the renal cystic disease
`are critical.
`Future clinical trials. The panel recommended that future
`clinical trials should include optimizing rapalog therapy by
`defining the minimum dose required that maintains a maximum
`response. Furthermore, identifying agents that could be com-
`bined with rapalogs or that could be used individually to induce
`a more complete and/or durable response could prove to be
`pivotal in allowing periodic or one-time, rather than life-long,
`therapy. In addition, determining whether catalytic mTOR
`inhibitors induce a more complete and/or durable response
`compared with rapalogs is a high priority.
`Prevention. In general, current studies have focused on the
`treatment of large or enlarging angiomyolipomas. Future stud-
`ies should focus on the prevention of angiomyolipomas, which
`would require additional natural history information, including
`defining whether there is a “window” of more rapid growth of
`angiomyolipomas during development. Although there is an
`anecdotal sense that angiomyolipomas can grow more rapidly
`during adolescence and early adulthood, this is not well de-
`fined. Epithelial cysts in TSC remain understudied, and the
`effects of rapalogs on cyst progression are unknown. Individ-
`uals with the TSC2/PKD1 contiguous gene syndrome, who are
`at risk for early-onset, severe polycystic kidney disease, rep-
`resent a population in whom prevention studies are a high
`priority.
`
`ACKNOWLEDGMENTS
`
`This work was submitted on behalf of the National Institute of Diabetes and
`Digestive and Kidney Diseases (NIDDK) TSC Working Group: E. Henske,
`Brigham and Women’s Hospital; J. Bissler, Le Bonheur Children’s Hospital;
`R. Rasooly, NIDDK; Chris Kingswood, Royal Sussex County Hospital,
`Brighton, UK; Elizabeth Thiele, Massachusetts General Hospital; Julian Samp-
`son, Institute of Medical Genetics, Cardiff University, Cardiff, UK; David
`Kwiatkowski, Harvard Medical School; Brendan Manning, Harvard School of
`Public Health; B. Siroky, Cincinnati Children’s Hospital Medical Center;
`Thomas Weimbs, University of California Santa Barbara; and Leon Murphy,
`Novartis Pharmaceuticals.
`
`DISCLOSURES
`
`No conflicts of interest, financial or otherwise, are declared by the authors.
`
`AJP-Renal Physiol • doi:10.1152/ajprenal.00525.2013 • www.ajprenal.org
`
`West-Ward Exhibit 1110
`Henske 2014
`Page 004
`
`

`

`Downloaded from
`
`http://ajprenal.physiology.org/
`
` by 10.220.33.4 on January 8, 2017
`
`TSC, mTOR, AND THE KIDNEY
`
`Review
`
`F283
`
`AUTHOR CONTRIBUTIONS
`
`Author contributions: E.P.H., B.J.S., and J.J.B. drafted manuscript; E.P.H.,
`R.R., B.J.S., and J.J.B. edited and revised manuscript; E.P.H., R.R., B.J.S., and
`J.J.B. approved final version of manuscript.
`
`REFERENCES
`
`1. Argani P, Aulmann S, Illei PB, Netto GJ, Ro J, Cho HY, Dogan S,
`Ladanyi M, Martignoni G, Goldblum JR, Weiss SW. A distinctive
`subset of PEComas harbors TFE3 gene fusions. Am J Surg Pathol 34:
`1395–1406, 2010.
`2. Bissler JJ, Kingswood JC. Renal angiomyolipomata. Kidney Int 66:
`924 –934, 2004.
`3. Bissler JJ, Kingswood JC, Radzikowska E, Zonnenberg BA, Frost M,
`Belousova E, Sauter M, Nonomura N, Brakemeier S, de Vries PJ,
`Whittemore VH, Chen D, Sahmoud T, Shah G, Lincy J, Lebwohl D,
`Budde K. Everolimus for angiomyolipoma associated with tuberous
`sclerosis complex or sporadic lymphangioleiomyomatosis (EXIST-2): a
`multicentre, randomised, double-blind, placebo controlled trial. Lancet
`381: 817–824, 2013.
`4. Bissler JJ, McCormack FX, Young LR, Elwing JM, Chuck G, Leon-
`ard JM, Schmithorst VJ, Laor T, Brody AS, Bean J, Salisbury S,
`Franz DN. Sirolimus for angiomyolipoma in tuberous sclerosis complex
`or lymphangioleiomyomatosis. N Engl J Med 358: 140 –151, 2008.
`5. Carsillo T, Astrinidis A, Henske EP. Mutations in the tuberous sclerosis
`complex gene TSC2 are a cause of sporadic pulmonary lymphangioleio-
`myomatosis. Proc Natl Acad Sci USA 97: 6085–6090, 2000.
`6. Dabora SL, Franz DN, Ashwal S, Sagalowsky A, DiMario FJ Jr, Miles
`D, Cutler D, Krueger D, Uppot RN, Rabenou R, Camposano S, Paolini
`J, Fennessy F, Lee N, Woodrum C, Manola J, Garber J, Thiele EA.
`Multicenter phase 2 trial of sirolimus for tuberous sclerosis: kidney
`angiomyolipomas and other tumors regress and VEGF- D levels decrease.
`PloS One 6: e23379, 2011.
`7. Dabora SL, Jozwiak S, Franz DN, Roberts PS, Nieto A, Chung J,
`Choy YS, Reeve MP, Thiele E, Egelhoff JC, Kasprzyk-Obara J,
`Domanska-Pakiela D, Kwiatkowski DJ. Mutational analysis in a cohort
`of 224 tuberous sclerosis patients indicates increased severity of TSC2,
`compared with TSC1, disease in multiple organs. Am J Hum Genet 68:
`64 –80, 2001.
`8. Davies DM, Johnson SR, Tattersfield AE, Kingswood JC, Cox JA,
`McCartney DL, Doyle T, Elmslie F, Saggar A, de Vries PJ, Sampson
`JR. Sirolimus therapy in tuberous sclerosis or sporadic lymphangioleio-
`myomatosis. N Engl J Med 358: 200 –203, 2008.
`9. Ewalt DH, Sheffield E, Sparagana SP, Delgado MR, Roach ES. Renal
`lesion growth in children with tuberous sclerosis complex. J Urol 160:
`141–145, 1998.
`10. Guo Y, Kwiatkowski DJ. Equivalent benefit of rapamycin and a potent
`mTOR ATP-competitive inhibitor, MLN0128 (INK128),
`in a mouse
`model of tuberous sclerosis. Mol Cancer Res 11: 467–473, 2013.
`11. Hartman TR, Liu D, Zilfou JT, Robb V, Morrison T, Watnick T,
`Henske EP. The tuberous sclerosis proteins regulate formation of the
`primary cilium via a rapamycin-insensitive and polycystin 1-independent
`pathway. Hum Mol Genet 18: 151–163, 2009.
`12. Henske EP, McCormack FX. Lymphangioleiomyomatosis—a wolf in
`sheep’s clothing. J Clin Invest 122: 3807–3816, 2012.
`
`13. Karbowniczek M, Yu J, Henske EP. Renal angiomyolipomas from
`patients with sporadic lymphangiomyomatosis contain both neoplastic and
`non-neoplastic vascular structures. Am J Pathol 162: 491–500, 2003.
`14. Krueger DA, Care MM, Holland K, Agricola K, Tudor C, Mangesh-
`kar P, Wilson KA, Byars A, Sahmoud T, Franz DN. Everolimus for
`subependymal giant-cell astrocytomas in tuberous sclerosis. N Engl J Med
`363: 1801–1811, 2010.
`15. McCormack FX, Inoue Y, Moss J, Singer LG, Strange C, Nakata K,
`Barker AF, Chapman JT, Brantly ML, Stocks JM, Brown KK, Lynch
`JP, 3rd Goldberg HJ, Young LR, Kinder BW, Downey GP, Sullivan
`EJ, Colby TV, McKay RT, Cohen MM, Korbee L, Taveira-DaSilva
`AM, Lee HS, Krischer JP, Trapnell BC. Efficacy and safety of sirolimus
`in lymphangioleiomyomatosis. N Engl J Med 364: 1595–1606, 2011.
`16. O’Brien K, Leach J, Jones B, Bissler J, Zuccarello M, Abruzzo T.
`Calcifications associated with pediatric intracranial arterial aneurysms:
`incidence and correlation with pathogenetic subtypes. Child’s Nerv Syst
`29: 643–649, 2013.
`17. Rakowski SK, Winterkorn EB, Paul E, Steele DJ, Halpern EF, Thiele
`EA. Renal manifestations of tuberous sclerosis complex: Incidence, prog-
`nosis, and predictive factors. Kidney Int 70: 1777–1782, 2006.
`18. Sampson JR, Maheshwar MM, Aspinwall R, Thompson P, Cheadle
`JP, Ravine D, Roy S, Haan E, Bernstein J, Harris PC. Renal cystic
`disease in tuberous sclerosis: role of the polycystic kidney disease 1 gene.
`Am J Hum Genet 61: 843–851, 1997.
`19. Sarbassov DD, Ali SM, Sengupta S, Sheen JH, Hsu PP, Bagley AF,
`Markhard AL, Sabatini DM. Prolonged rapamycin treatment inhibits
`mTORC2 assembly and Akt/PKB. Mol Cell 22: 159 –168, 2006.
`20. Serra AL, Poster D, Kistler AD, Krauer F, Raina S, Young J, Rentsch
`KM, Spanaus KS, Senn O, Kristanto P, Scheffel H, Weishaupt D,
`Wuthrich RP. Sirolimus and kidney growth in autosomal dominant
`polycystic kidney disease. N Engl J Med 363: 820 –829.
`21. Shillingford JM, Murcia NS, Larson CH, Low SH, Hedgepeth R,
`Brown N, Flask CA, Novick AC, Goldfarb DA, Kramer-Zucker A,
`Walz G, Piontek KB, Germino GG, Weimbs T. The mTOR pathway is
`regulated by polycystin-1, and its inhibition reverses renal cystogenesis in
`polycystic kidney disease. Proc Natl Acad Sci USA 103: 5466 –5471,
`2006.
`22. Vollenbroker B, George B, Wolfgart M, Saleem MA, Pavenstadt H,
`Weide T. mTOR regulates expression of slit diaphragm proteins and
`cytoskeleton structure in podocytes. Am J Physiol Renal Physiol 296:
`F418 –F426, 2009.
`23. Walz G, Budde K, Mannaa M, Nurnberger J, Wanner C, Sommerer
`C, Kunzendorf U, Banas B, Horl WH, Obermuller N, Arns W,
`Pavenstadt H, Gaedeke J, Buchert M, May C, Gschaidmeier H,
`Kramer S, Eckardt KU. Everolimus in patients with autosomal dominant
`polycystic kidney disease. N Engl J Med 363: 830 –840.
`24. Williams JM, Racadio JM, Johnson ND, Donnelly LF, Bissler JJ.
`Embolization of renal angiomyolipomata in patients with tuberous scle-
`rosis complex. Am J Kidney Dis 47: 95–102, 2006.
`25. Young LR, Vandyke R, Gulleman PM, Inoue Y, Brown KK, Schmidt
`LS, Linehan WM, Hajjar F, Kinder BW, Trapnell BC, Bissler JJ,
`Franz DN, McCormack FX. Serum vascular endothelial growth factor-D
`prospectively distinguishes lymphangioleiomyomatosis from other dis-
`eases. Chest 138: 674 –681, 2010.
`
`AJP-Renal Physiol • doi:10.1152/ajprenal.00525.2013 • www.ajprenal.org
`
`West-Ward Exhibit 1110
`Henske 2014
`Page 005
`
`

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