throbber
Molecular-based Therapies
`for Renal Cell Carcinoma
`Amnon Zisman, MD, Allan J. Pantuck, MD,
`and Arie S. Belldegrun, MD, FACS
`
`Address
`Department of Urology, University of California School of
`Medicine, Le Conte Avenue, Room 66-118 CHS, Los Angeles,
`CA 90095-1738, USA.
`E-mail: tprintz@mednet.ucla.edu
`Current Urology Reports 2001, 2:55–61
`Current Science Inc. ISSN 1527–2737
`Copyright © 2001 Current Science Inc.
`
`The incidence of renal cell carcinoma (RCC) is rising
`steadily, but the ability to cure patients with metastatic
`RCC unfortunately remains limited. Emerging interest in
`gene therapy performance and safety is expressed by
`patients, medical institutes, and other agencies. It has
`become evident that better understanding of the genetic
`impairments and immune pathophysiology in RCC is
`essential for future improvement in patient care. Clinical
`trials now underway that are focusing on genetic and
`immune impairments will hopefully lead to future break-
`throughs in RCC therapy. This paper reviews available gene
`therapies and other related therapeutic approaches for
`RCC and lists some of the current clinical trials focused
`on molecular-based therapies.
`
`Introduction
`Renal cell carcinoma (RCC) is the most common tumor
`rising from the kidney. Approximately 30,000 cases
`per year are diagnosed in the United States, and incidence
`is increasing [1]. RCC is frequently resistant to radiation
`and chemotherapy, leaving surgical removal of the tumor
`the treatment of choice for localized renal cell cancer.
`However, patients with a disseminated disease have a poor
`prognosis, with an average survival of less than 1 year from
`the time of diagnosis [2]. Traditionally, nephrectomy is not
`usually performed when metastases are present, and the
`objective response rate to conventional chemotherapy is
`only 6% [3]. About 1% to 5% of patients with RCC have
`spontaneous regression, mainly of their pulmonary
`metastases [4]. The reason for this is unknown but is gener-
`ally attributed to self-immune mechanisms. With this in
`mind, methods are being developed to boost the human
`immune system to fight metastatic tumors (Fig. 1). These
`methods are generally termed immunotherapy.
`
`Immunotherapy
`The rationale for turning the immune system against tumor
`cells is based on the theory that the tumor cells survive
`through bypassing and inhibition of immune surveillance
`mechanisms (eg, by downregulation of cell surface antigens,
`such as the major histocompatibility complex [5] and by local
`hindrance of tumor-infiltrating immune competent cells).
`The first attempts to reinforce immunomodulation included
`direct systemic administration of nonspecific stimulants, such
`as the cytokines IL-2, interferon-a (IFN-a), IL-4, GM-CSF, and
`IL-12. Alternatively, adoptive (passive) immunotherapy was
`applied, in which self-lymphocytes were exposed ex vivo to
`the cytokines and then reinjected to the patient. The first
`attempts to apply adoptive immunotherapy used nonspecific
`LAK cells that were isolated from the patient’s peripheral
`blood, incubated in vitro with IL-2, and reinfused into the
`patient. A number of clinical trials demonstrated an average
`response rate of 23.5 % [6]. A more specific application is the
`combination of tumor-infiltrating leukocytes (TILs), which
`are isolated from the tumor tissue and are assumed to have
`immunologic memory to tumor antigens. On a cellular
`basis, TILs are 50 to 100 times more potent than LAK cells in
`mediating tumor regression. Few clinical studies have been
`undertaken with TILs. At the University of California, Los
`Angeles (UCLA), TILs are isolated from fresh nephrectomy
`specimens of RCC patients, expanded in vitro with IL-2 in the
`presence of tumor extract, and reinfused to the patient. The
`response rate obtained with this protocol was 33%, and the
`average response duration was 14 months, with a mean sur-
`vival of 22 months [7]. Recently, more tumor-specific
`approaches have been developed, targeting specific tumor
`markers in an attempt to limit the immune destruction to the
`malignant cells. These techniques are tailored to each patient.
`One promising example is the use of autologous dendritic
`cells (DCs) that were exposed ex vivo to specific tumor anti-
`gens. These DCs serve as antigen-presenting cells that present
`the tumor epitopes to T cells in situ, leading to anti-tumor
`immunity. This approach is currently being tested in a
`clinical trial at UCLA [8].
`Lately, preliminary data on two exciting new approaches
`have been reported showing impressive rates of regression
`of metastatic foci in patients with advanced RCC. These
`studies extend and improve the dendritic cell tumor vaccine
`approach by using an autologous tumor cell electrically
`
`NOVARTIS EXHIBIT 2097
`Breckenridge v. Novartis, IPR 2017-01592
`Page 1 of 7
`
`

`

`56
`
`New Techniques, Molecular Gene Therapy
`
`CTL
`
`.........
`
`-cell
`medlailecl
`tlJI
`
`, ...
`
`/
`
`1d UC
`
`Figure 1. Schematic representation of potential molecular-based therapies for renal cell carcinoma. AP—antigen presenting;
`CTL—cytotoxic T cell; DC—dendritic cell; WT—wild type.
`
`fused with allogenic dendritic cells to form an RCC-DC
`hybrid cell [9••]. Similarly interesting reports on the use
`of nonmyeloablative allogeneic peripheral-blood stem-cell
`transplantation give hope for patients failing currently
`existing immunotherapies [10•].
`Cytokine-based immunotherapy has several advantages:
`1) it is easy to produce large quantities by recombinant DNA
`technology; 2) administration of this immunotherapy does
`not require complicated preparation procedures; and 3)
`FDA approval of a universal therapeutic agent, such as a
`cytokine, is more straightforward than a patient-dependent
`“tailor-made” treatment protocol. As a result, IL-2 has
`become an FDA-approved treatment for metastatic RCC in a
`relatively short period of time. Although interferon-based
`therapy generates similar overall response rates to IL-2 (15%
`to 20%) [6], high-dose IL-2 monotherapy causes a higher
`frequency of complete remission and prolonged response
`durations. Regardless of the specific immunotherapy regi-
`men used, patients included in immunotherapy clinical
`trials had better prognosis than patients treated by other
`clinical protocols in which no immunotherapy was given
`[11]. Some common protocols for IL-2 immunotherapy and
`their outcome are listed in Table 1 [12]. The overall outcome
`of high-dose intravenous recombinant interleukin (rIL-2)
`alone appears to be beneficial to each of the other regimens.
`However, this treatment is accompanied by very high toxic-
`ity and other adverse reactions [13]. Therefore, other strate-
`gies for therapy are continuously tested.
`
`Molecular-based Therapeutic Strategies
`Molecular-based therapeutic strategies have evolved beyond
`the previously described immunotherapeutic techniques to
`
`include a variety of genes and cell signaling pathways.
`Classification of these therapies, based on their strategic
`approach to eliminate tumor cells, is listed in Table 2.
`
`Immune-based gene therapy (cancer vaccines)
`This therapeutic approach aims to preserve the anti-tumor
`activity of cytokines while minimizing their systemic
`adverse effects. In this approach, cytokine genes or growth
`factor genes are introduced locally by various transfection
`and infection techniques, aiming at confined production
`of high levels of the gene products within the tumor vicin-
`ity. The gene products that are produced locally in high
`concentrations may directly alter neoplastic properties
`associated with invasion and metastasis. This approach
`carries significant advantage in that it is associated with
`lower toxicity than systemic administration of cytokines
`[14]. Animal studies using this technique have demon-
`strated prevention of tumor growth, decreased metastatic
`spread, and prolonged immunologic memory, resulting
`in rejection of subsequent tumor challenges. At UCLA we
`have transfected the genes for IL-2 and IFN-a into human
`RCC lines. When these cells were implanted subcutane-
`ously into nude mice, their cytokine secretion inhibited
`local tumor growth in a more effective manner than
`systemic administration of IL-2 and IFN-a [15]. In addi-
`tion to IL-2 and IFN-a, various other genes of immune
`system modulators, including GM-CSF [16] and B7
`(a costimulatory molecule needed for T cell activation
`[17]), were tested as possible tumor vaccines for RCC [18].
`One of the options used to confine the cytokines into the
`tumor site is by transfecting tumor infiltrating lymphocytes
`(TILs) with the cytokine genes [19]. The rationale is
`that TILs will home back to tumor deposits. In this way,
`
`NOVARTIS EXHIBIT 2097
`Breckenridge v. Novartis, IPR 2017-01592
`Page 2 of 7
`
`

`

`Molecular-based Therapies for Renal Cell Carcinoma • Zisman et al.
`
`57
`
`Table 1. Protocols tested in interleukin-2 immunotherapy for renal cell carcinoma
`
`Regimen
`
`Response rate, %
`
`Complete
`response rate, %
`
`Median response
`duration, mo
`
`High-dose rIL-2 alone
`High-dose rIL-2/rIFN-a
`Outpatient subcutaneous rIL-2/rIFN-a
`Outpatient rIL-2/rIFN-a, plus 5-FU/rIFN-a
`
`17
`11
`17
`16
`
`7
`0
`4
`4
`
`53
`7
`12
`9
`
`Table 2. Classification of molecular-based therapies for renal cell carcinoma
`
`Immunotherapy
`Cytokine administration (eg, IL-2, interferon, IL-4, GM-CSF, IL-12)
`Adoptive immunotherapy: ex vivo activation of autologous immune effector cells using recombinant cytokines followed
`by reinfusion to the patient
`Antigen-presenting cells therapy alone or in combination with cytokines (dendritic cell vaccines)
`Immune-based gene therapy (cancer vaccines)
`Cells transfected with cytokine genes or growth factors genes (IL-2, GM-CSF [G-VAX], IL-12) are introduced to the tum
`or site and locally release the transfected gene product, inducing immune activation with lower systemic toxicity than
`with direct injection of cytokines. The transfected cells can be autologous tumor cells or TILs.
`Viruses as gene carriers (adeno viruses, retro viruses, pox, and vaccinia)
`Direct transfection with DNA/genes
`Cytoreductive therapies
`Suicide genes (eg, thymidine kinase gene therapy followed by gancyclovir administration)
`Drug-activated suicide genes
`Oncolytic viruses (eg, adenovirus replicating in p53 deficient cells only)
`Toxic gene therapy (necrosis and apoptosis induction by diphteria toxin)
`Corrective gene therapy
`Introduction of active wild type suppressor genes (eg, p53, VHL gene, p16, p27) into tumor cells with defective or
`inactive tumor suppressor genes
`Tumor growth and survival factor modulation by antisense mRNA molecules (eg, antisense bcl-2 and antisense TGF-b)
`
`secretion of the relevant gene product may be localized
`and continuous, leading to augmented killing. A phase I
`clinical trial using irradiated RCC cells transfected ex vivo
`with human GM-CSF gene was performed to test the safety
`and the induction of immune responses in patients with
`metastatic RCC [20]. No significant toxicity or autoim-
`mune responses were reported, and one of 16 patients had
`a partial response. Furthermore, recent studies using modi-
`fied dendritic cells [21] and studies directly introducing the
`cytokines into the tumor have been performed at UCLA as
`well as in other centers. At this time, tumor vaccine-based
`gene therapy appears to be safe, but its efficacy in meta-
`static RCC has yet to be proven.
`
`Cytoreductive therapy
`This therapeutic line shares the concept of introducing
`specific genes into the tumor site. However, in cyto-
`reductive therapy, a suicidal gene is introduced into the
`tumor. In most cases, the gene encodes for an enzyme
`capable of converting an otherwise benign medication
`(pro-drug) into a highly cytotoxic one. Administration
`of the pro-drug produces a high concentration of the cyto-
`toxic metabolite in the tumor area, but not in normal
`tissues that did not encounter the gene, resulting in
`reduced systemic toxicity. The most commonly used
`system in this group is herpes simplex thymidine kinase
`
`gene (HSV-tk). HSV-tk phosphorylates ganciclovir to ganci-
`clovir monophosphate, which is converted to ganciclovir
`triphosphate by cellular kinases. The resulting triphos-
`phate acts as a false base, inhibiting DNA polymerase and
`DNA synthesis, leading to cell death. Other examples of
`cytoreductive strategies include virus systems that target
`only p53 deficient cells and the diphtheria toxin gene,
`whose product induces necrosis and apoptosis.
`
`Corrective gene therapy
`This therapeutic approach attempts to correct a specific
`genetic alteration that was found to be present in a certain
`malignancy, such as overexpression of an oncogene or
`inactivation of a tumor suppressor gene (eg, p53 gene) by a
`mutation. The logic for corrective gene therapy is that the
`rectification should restore normal growth control. Correc-
`tive gene therapy uses the same vectors as immunotherapy
`and cytoreductive therapy to introduce the relevant wild
`type gene into the tumor cells. Alternatively, corrective
`gene therapy can be achieved by introducing the comple-
`mentary stretch of an mRNA, whose further translation to
`an active protein is undesired. This complementary stretch,
`called “antisense mRNA” binds to the sense-mRNA and
`blocks the translation of its protein. This was experimented
`with oncogenes like bcl-2 and growth factor genes like
`transforming growth factor (TGF) b [22].
`
`NOVARTIS EXHIBIT 2097
`Breckenridge v. Novartis, IPR 2017-01592
`Page 3 of 7
`
`

`

`58
`
`New Techniques, Molecular Gene Therapy
`
`Tumorigenetic Pathways
`of Renal Cell Carcinoma:
`Mechanisms and Therapeutic Implications
`Chromosome 3 and von Hippel-Lindau gene
`Genetic studies of families with hereditary RCC, in
`the presence or absence of von Hippel-Lindau (VHL)
`syndrome, contributed to our current view of the tumori-
`genetic pathways of RCC, possibly leading to the design
`of future gene-targeted therapies. These studies identified
`a loss of chromosome 3p in many sporadic and familial
`RCC patients [23] and localized the VHL gene on chro-
`mosome 3p25-26 [23,24]. The wild type (WT) VHL gene
`product, pVHL, is a 213 amino acid polypeptide that
`forms multiprotein complexes with elongin B, elongin C,
`and Cul-2 and negatively regulates hypoxia-inducible
`mRNAs, such as the mRNA encoding vascular endothelial
`growth factor (VEGF). pVHL is suspected to play a role in
`ubiquitination given the similarity of elongin C and Cul-
`2 with Skp1 and Cdc53, respectively. Furthermore, pVHL
`can interact with fibronectin and is required for the
`assembly of a fibronectin matrix. Thus, pVHL is a tumor
`suppressor protein that regulates angiogenesis, extracellu-
`lar matrix formation, and the cell cycle [25]. More than
`half of the patients with sporadic RCC have a detectable
`mutation in one allele of the VHL gene [26], and allelic
`loss in the other allele is seen in up to 98% of tumors
`[27,28]. Gross karyotypic changes of chromosome 3,
`such as chromosomal 3p loss or monosomy, were also
`observed in some patients [29,30]. Moreover, subtle
`molecular changes in the form of hypermethylation of
`the DNA in regulatory areas of the VHL gene, which were
`found in up to 20% of sporadic RCC [31,32], caused tran-
`scriptional arrest. Additional gene loci on chromosome
`3p [33,34] and aberrations in chromosomes 5, 7, 14, and
`Y [35] were also associated with RCC. These loci may be
`able to act independently from the VHL locus, resulting
`in the development of RCC. Moreover, it was recently
`shown that the loss of VHL gene is associated with
`increased expression of the inhibitory cytokines TGFa
`and b1 [36•]. This ties together the genetic and immuno-
`logic alterations seen with RCC.
`Therefore, the VHL gene and gene products may serve
`as potential targets for corrective gene therapy. Initial
`studies have been performed to replace the defective tumor
`suppressor product by the WT gene in an attempt to reverse
`the cancer phenotype. The WT VHL gene was transfected
`into RCC cell lines that lacked the normal expression of the
`gene, attached to a constitutively activated cytomegalovirus
`promoter via a liposome vehicle [37••]. Transfection of the
`WT VHL gene resulted in growth suppression of the RCC
`cell line. In contrast to cultures of mutant VHL RCC cells,
`which formed very compact and cohesive spheroids, the
`WT VHL transfectants were loosely arranged and formed a
`network of tubular and trabecular structures within the
`spheroids [38]. The morphologic changes of the WT VHL
`spheroids were associated with the deposition of fibronec-
`
`tin in the extracellular space, a feature that was absent in the
`mutant and inactivated VHL gene-expressing spheroids.
`The results suggest that the VHL gene may be involved in
`the maintenance of the epithelial phenotype of renal tubu-
`lar cells, ie, it may act as a differentiation/morphogenetic
`factor. Moreover, this effect in tumor cells appears to be
`highly dependent on multicellular growth conditions that
`mimic the basic nature of solid tumors, such as RCC [38].
`In addition, it was recently shown that loss of VHL gene was
`associated with increased expression of TGF-a and TGF-b1,
`and that transfection of RCC cell lines with WT VHL sub-
`stantially decreased TGF-a and TGF-b1 mRNA and protein
`by shortening their mRNA half-life [36,39]. Thus, gene
`replacement therapy using the WT VHL gene may have a
`role in treating patients with RCC, although the safety and
`efficacy of this treatment is yet to be defined.
`
`P53 mutation
`p53 was named Molecule of the Year in 1993 by the editors
`of Science [40], and the “guardian of the genome” by many
`researchers. The human p53 tumor suppressor gene is
`located on chromosome 17p13.1 and encodes a nuclear
`transcription factor that has been implicated in normal
`development and cellular responses to stress. The WT protein
`plays a major role in DNA transcription, cell growth and pro-
`liferation, and a number of metabolic processes. It
`suppresses abnormal cell proliferation and is involved in
`programmed cell death, or apoptosis in damaged cells
`[41,42]. The mechanisms of growth arrest and apoptosis are
`both direct, intracellular effects moderated by p53-respon-
`sive genes and bystander, intercellular mechanisms involving
`secretion of growth inhibitory factors [43]. Mutations in the
`p53 gene lead to loss of the tumor suppressor capabilities
`and to cancer promotion. A large number of human cancers
`contain p53 mutations, including cancers of the breast, cer-
`vix, colon, lung, liver, prostate, bladder, and skin. In RCC,
`p53 is associated with the aggressive sarco-matoid variant
`(79%) [44]. The loss of p53 function in these tumors is asso-
`ciated with more aggressiveness, higher frequency of
`metastasis, and resistance to anti-cancer therapy.
`The WT p53 suppresses progression through the cell cycle
`in response to DNA damage, thereby allowing DNA repair to
`occur before replicating the genome. Therefore, p53 prevents
`the transmission of damaged genetic information from one
`cell generation to the next and initiates apoptosis if the
`damage to the cell is severe. The carboxy-terminus of human
`p53 plays an important role in controlling the specific DNA
`binding function. WT p53 is found in a latent form, which
`does not bind to DNA. The specific DNA binding activity was
`shown to be activated by various pathways: phosphorylation,
`antibody specific for the carboxy-terminus of the protein,
`small peptides that could mimic the carboxy-terminus of the
`p53, short single-stranded DNA, deletion of the last
`30 amino acids, and the interaction with a cellular protein
`[45–47]. Thus, all naturally occurring mutations in p53
`directly or indirectly affect the interaction of p53 with DNA,
`
`NOVARTIS EXHIBIT 2097
`Breckenridge v. Novartis, IPR 2017-01592
`Page 4 of 7
`
`

`

`Molecular-based Therapies for Renal Cell Carcinoma • Zisman et al.
`
`59
`
`Table 3. Current clinical trials using molecular based therapies for advanced renal cell carcinoma
`
`Basic principle
`
`Principal investigator
`
`Intratumoral injection of LEUVECTIN (phase II)
`TIL+INF+IL-2
`Liposome IL-2
`HLA-B7 and IL-2 gene
`Multi-antigen loaded dendritic cell vaccine
`(Adoptive immunotherapy - phase I)
`HLA-B7 and IL-2
`IL-4
`TNF- a
`IL-2
`Liposome HLA-B7/b-2 microglobulin
`Autologous tumor cell vaccine+IFN/GM-CSF (phase II)
`IL-2 (allogeneic)
`GM-CSF
`HLA-B7/b-2 microglobulin
`
`Belldegrun
`Belldegrun
`Figlin
`Figlin
`Gitlitz
`
`Antonia
`Lotze
`Rosenberg
`Rosenberg
`Chang
`Dillman
`Gansbacher
`Simons
`Fox
`
`Site
`
`UCLA
`UCLA
`UCLA
`UCLA
`UCLA
`
`University of South Florida
`University of Pittsburgh
`NIH
`NIH
`multicenter
`multicenter
`MSKC
`Johns Hopkins
`Chiles Research Institute
`
`Adapted with changes from the National Cancer Institute web site: www.cancernet.mci.nih.gov and from Rodriguez et al. [58].
`MSKC—Memorial Sloan-Kettering Cancer Center; NIH—National Institutes of Health; UCLA—University of California, Los Angeles.
`
`demonstrating that sequence-specific DNA binding is central
`to the normal functioning of p53 as a tumor suppressor.
`Therefore, p53 gene and gene products may serve
`as potential targets for corrective gene therapy. In vitro
`attempts to correct the p53 gene in RCC cell lines using
`liposome-p53 gene complexes have resulted in decreased
`growth of tumor cells in culture. Transfection of the p53
`gene into a mouse xenograft model resulted in a decrease in
`the number of metastatic lung lesions [48]. A phase I dose
`escalation study of a single intratumoral injection of a
`replication-defective adenoviral expression vector encoding
`WT p53 was carried out in patients with incurable non-
`small cell lung cancer. All patients enrolled had p53 protein
`overexpression as a marker of mutant p53 status in pretreat-
`ment tumor biopsies. No clinically significant toxicity
`was observed; however, successful transfer of WT
`p53 was achieved only with higher vector doses [49]. In a
`recent phase I study, patients with non-small cell lung
`cancer received repeated intratumoral injections of Ad-p53.
`Repeated injections appear to be equally tolerated, resulted
`in transgene expression of WT p53, and seemed to mediate
`antitumor activity in a subset of patients [50]. The use of
`the p53 WT gene therapy by intratumoral injection may
`prove effective for additional tumors.
`
`G250
`G250 is a tumor marker recently introduced for RCC. This
`protein is detected on most primary and metastatic RCCs
`but is absent from kidney and other normal tissues,
`with the exception of gastric mucosal cells and cells of the
`larger bile ducts [50,52]. G250 is a transmembrane protein
`identical to the tumor-associated antigen MN/CA IX that
`was previously identified in cervical carcinoma. It is a
`carbonic anhydrase IX tumor antigen and is expressed only
`when a mutation occurs either within the coding region of
`
`the elonging binding domain of the VII gene, or when a 5’
`non-sense or frameshift mutation occurs [53]. However, its
`unique expression on RCC tissues makes it a potential can-
`didate for both tumor diagnosis and therapy [54,55]. Vari-
`ous potential therapeutic approaches can be considered,
`including targeting the tumors by specific anti-G250 anti-
`bodies or educating T cells to specific killing by presenting
`them with G250 epitopes using dendritic cell tumor vac-
`cines [56•]. Further studies are currently ongoing to eluci-
`date its potential role. At UCLA we fused the DNA
`sequences of the G250 renal cell carcinoma-associated
`antigen together with granulocyte-macrophage colony
`stimulating factor (GM-CSF) and cloned a novel G250-
`GM-CSF fusion protein [57]. The fusion protein retains
`its GM-CSF biologic activity and together with IL-4 is
`capable of inducing dendritic cell (DC) differentiation.
`The activated DCs induce specific killing activity by cyto-
`toxic T cells against allogeneic tumors cultured from meta-
`static RCC patients. This new strategy opens multiple
`therapeutic possibilities and may obviate fresh tumor spec-
`imen for the generation of anti-tumor vaccines. It may be
`possible that patients’ DCs will be activated with the fusion
`protein and, together with systemic IL-2 administration,
`will activate tumor-infiltrating lymphocytes specifically
`against G250 on RCC cells. A list of clinical trials using
`molecular-based therapies is presented (Table 3).
`
`Conclusions
`In the past 20 years, impressive advances in the application
`of immunotherapy to treating renal cell carcinoma were
`witnessed. At UCLA we have seen a progressive increase
`in responses to treatment as therapy has evolved, from
`systemic IFNa administration (16%), to combination
`IFN+IL-2 (25%), to the current method of bulk TILs (33%)
`
`NOVARTIS EXHIBIT 2097
`Breckenridge v. Novartis, IPR 2017-01592
`Page 5 of 7
`
`

`

`60
`
`New Techniques, Molecular Gene Therapy
`
`and CD8-/TILs (40%). Patient characteristics that predict
`improved responsiveness to therapy have been identified,
`and treatment protocols that decrease toxicity have been
`developed. The most encouraging results have been the
`improved rates of complete clinical response, most of
`which are durable and long-lasting.
`There is no doubt that current immunotherapeutic
`protocols produce changes in the natural history of this
`disease and cause significant and lasting remissions in
`select patients. Moreover, the place of surgery in advanced
`RCC is no longer anecdotal. With the advantages of tumor-
`infiltrating leukocyte technology, nephrectomy is essential
`for the preparation of tumor-infiltrating leukocytes.
`
`References and Recommended Reading
`Papers of particular interest, published recently, have been
`highlighted as:
`•
`Of importance
`•• Of major importance
`
`6.
`
`7.
`
`2.
`
`3.
`
`4.
`
`1. Chow WH, Devesa SS, Warren JL, Fraumeni JFJ: Rising
`incidence of renal cell cancer in the United States.
`JAMA 1999, 281:1628–1631.
`Elson PJ, Witte RS, Trump DL: Prognostic factors for survival
`in patients with recurrent or metastatic renal cell carcinoma.
`Cancer Res 1988, 48:7310–7313.
`Yagoda A, Abi-Rached B, Petrylak D: Chemotherapy for
`advanced renal-cell carcinoma: 1983–1993. Semin Oncol
`1995, 22:42–60.
`Papac R: Spontaneous regression of cancer. Cancer Treat Rev
`1996, 22:395–423.
`5. Garrido F, Ruiz-Cabello F: MHC expression on human
`tumors—its relevance for local tumor growth and metastasis.
`Semin Cancer Biol 1991, 2:3–10.
`Bukowski RM: Natural history and therapy of metastatic renal
`cell carcinoma: the role of interleukin-2. Cancer
`1997, 80:1198–1220.
`Pierce WC, Belldegrun A, Figlin RA: Cellular therapy: scientific
`rationale and clinical results in the treatment of metastatic
`renal-cell carcinoma. Semin Oncol 1995, 22:74–80.
`8. Gitlitz B, Hinkel A, Mulders P, et al.: Multi-antigen loaded
`dendritic cell (DC) vaccine for the treatment of metastatic
`renal cell carcinoma (mRCC)-in-vitro correlates.
`J Urol 1999, 161:137a.
`9.•• Kugler A, Stuhler G, Walden P, et al.: Regression of human
`metastatic renal cell carcinoma after vaccination with
`tumor cell-dentric cell hybrids. Nat Med 2000, 6:332–336.
`An excellent description of a milestone in the evolution of
`modern molecular-based therapies for metastatic RCC. This
`study sets the direction for dendritic cell tumor vaccines and
`novel molecule targeting.
`10.• Childs R, Chernoff A, Contentin N, et al.: Regression of
`metastatic renal cell carcinoma after nonmyeloablative
`allogeneic peripheral blood stem-cell transplantation.
`N Engl J Med 2000, 343:750–757.
`An excellent description of a milestone in the evolution of modern
`molecular-based therapies for metastatic RCC.
`11. Motzer RJ, Mazumdar M, Bacik J, et al.: Survival and
`prognostic stratification of 670 patients with advanced
`renal cell carcinoma. J Clin Oncol 1999, 17:2530–2540.
`12. Dutcher JP, Atkins M, Fisher R, et al.: Interleukin-2-based
`therapy for metastatic renal cell cancer: the cytokine
`working group experience, 1989-1997. Cancer J Sci Am
`1997, 3 (suppl1):S73–S78.
`13. Wirth M: Immunotherapy for metastatic renal cell carcinoma.
`Urol Clin North Am 1993, 20:283–295.
`
`14.
`
`15.
`
`16.
`
`17.
`
`20.
`
`Saffran DC, Horton HM, Yankauckas MA, et al.: Immuno-
`therapy of established tumors in mice by intratumoral
`injection of interleukin-2 plasmid DNA: induction of
`CD8+ T-cell immunity. Cancer Gene Ther 1998, 5:321–330.
`Belldegrun A, Tso CL, Sakata T, et al.: Human renal carcinoma
`line transfected with interleukin-2 and/or interferon alpha
`gene(s): implications for live cancer vaccines.
`J Natl Cancer Inst 1993, 85:207–216.
`Seigne J, Turner J, Diaz J, et al.: Feasibility study of gene
`gun mediated immunotherapy for renal cell carcinoma.
`J Urol 1999, 162:1259–1263.
`Jung D, Hilmes C, Knuth A, et al.: Gene transfer of the
`co-stimulatory molecules B7-1 and B7-2 enhances the
`immunogenicity of human renal cell carcinoma to a
`different extent. Scand J Immunol 1999, 50:242–249.
`18. Gitlitz BJ, Belldegrun A, Figlin RA: Immunotherapy and
`gene therapy. Semin Urol Oncol 1996, 14:237–243.
`19. Mulders P, Tso CL, Pang S, et al.: Adenovirus-mediated
`interleukin-2 production by tumors induces growth of
`cytotoxic tumor-infiltrating lymphocytes against human
`renal cell carcinoma. J Immunother 1998, 21:170–180.
`Simons JW, Jaffee EM, Weber CE, et al.: Bioactivity of
`autologous irradiated renal cell carcinoma vaccines
`generated by ex vivo granulocyte-macrophage colony-
`stimulating factor gene transfer. Cancer Res
`1997, 57:1537–1546.
`21. Mulders P, Tso CL, Gitlitz B, et al.: Presentation of renal
`tumor antigens by human dendritic cells activates tumor-
`infiltrating lymphocytes against autologous tumor:
`implications for live kidney cancer vaccines.
`Clin Cancer Res 1999, 5:445–454.
`22. Morelli S, Alama A, Quattrone A, et al.: Oligonucleotides
`induce apoptosis restricted to the t(14;18) DHL-4 cell
`line. Anticancer Drug Des 1996, 11:1–14.
`23. Glenn GM, Linehan WM, Hosoe S, et al.: Screening for von
`Hippel-Lindau disease by DNA polymorphism analysis.
`JAMA 1992, 267:1226–1231.
`Linehan WM, Lerman MI, Zbar B: Identification of the von
`Hippel-Lindau (VHL) gene. Its role in renal cancer. JAMA
`1995, 273:564–570.
`25. Ohh M, Kaelin WJ: The von Hippel-Lindau tumour
`suppressor protein: new perspectives. Mol Med Today
`1999, 5:257–263.
`Shuin T, Kondo K, Torigoe S, et al.: Frequent somatic
`mutations and loss of heterozygosity of the von
`Hippel-Lindau tumor suppressor gene in primary human
`renal cell carcinomas. Cancer Res 1994, 54:2852–2855.
`27. Gnarra JR, Tory K, Weng Y, et al.: Mutations of the VHL
`tumour suppressor gene in renal carcinoma. Nat Genet
`1994, 7:85–90.
`Anglard P, Tory K, Brauch H, et al.: Molecular analysis of
`genetic changes in the origin and development of renal
`cell carcinoma. Cancer Res 1991, 51:1071–1077.
`Kovacs G, Emanuel A, Neumann HP, Kung HF: Cytogenetics
`of renal cell carcinomas associated with von Hippel-Lindau
`disease. Genes Chromosomes Cancer 1991, 3:256–262.
`Foster K, Prowse A, van den Berg A, et al.: Somatic mutations
`of the von Hippel-Lindau disease tumour suppressor gene in
`non-familial clear cell renal carcinoma. Hum Mol Genet
`1994, 3:2169–2173.
`31. Glavac D, Ravnik-Glavac M, Ovcak Z, Masera A: Genetic
`changes in the origin and development of renal cell
`carcinoma (RCC). Pflugers Arch 1996, 431:R193–R194.
`32. Herman JG, Latif F, Weng Y, et al.: Silencing of the VHL
`tumor-suppressor gene by DNA methylation in renal
`carcinoma. Proc Natl Acad Sci U S A 1994, 91:9700–9704.
`Foster K, Crossey PA, Cairns P, et al.: Molecular genetic
`investigation of sporadic renal cell carcinoma: analysis
`of allele loss on chromosomes 3p, 5q, 11p, 17 and 22.
`Br J Cancer 1994, 69:230–234.
`
`24.
`
`26.
`
`28.
`
`29.
`
`30.
`
`33.
`
`NOVARTIS EXHIBIT 2097
`Breckenridge v. Novartis, IPR 2017-01592
`Page 6 of 7
`
`

`

`Molecular-based Therapies for Renal Cell Carcinoma • Zisman et al.
`
`61
`
`36.
`
`35.
`
`34. Ohta M, Inoue H, Cotticelli MG, et al.: The FHIT gene,
`spanning the chromosome 3p14.2 fragile site and renal
`carcinoma-associated t(3;8) breakpoint, is abnormal in
`digestive tract cancers. Cell 1996, 84:587–597.
`Iqbal MA, Akhtar M, Ali MA: Cytogenetic findings in renal
`cell carcinoma. Hum Pathol 1996, 27:949–954.
`Knebelmann B, Ananth S, Cohen HT, Sukhatme VP: Trans-
`forming growth factor alpha is a target for the von Hippel-
`Lindau tumor suppressor. Cancer Res 1998, 58:226–231.
`37. Chen F, Kishida T, Duh FM, et al.: Suppression of growth
`of renal carcinoma cells by the von Hippel-Lindau tumor
`suppressor gene. Cancer Res 1995, 55:4804–4807.
`38.• Lieubeau-Teillet B, Rak J, Jothy S, et al.: von Hippel-Lindau
`gene-mediated growth suppression and induction of
`differentiation in renal cell carcinoma cells grown as multi-
`cellular tumor spheroids. Cancer Res 1998, 58:4957–4962.
`An excellent description of a milestone in the evolution of modern
`molecular-based therapies for metastatic RCC.
`39.•• Ananth S, Knebelmann B, Grüning W, et al.: Transforming
`growth factor beta1 is a target for the von Hippel-Lindau
`tumor suppressor and a critical growth fact

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