throbber
International Journal of Urology (2001) 8, S1–S4
`
`Conference Proceedings
`
`Gene and immune therapy for renal cell carcinoma
`
`ALLAN J PANTUCK, AMNON ZISMAN AND ARIE BELLDEGRUN
`Division of Urologic Oncology, Department of Urology, University of California School of
`Medicine, Los Angeles, California, USA
`
`Abstract
`
`Conventional therapy for metastatic renal cell carcinoma is associated with a poor response rate
`and few patients are long-term survivors. The occurrence of spontaneous regression and the pro-
`longed latency period between primary tumor removal and the appearance of metastases in some
`patients suggest the existence of important host immune responses to autologous tumor cells. With
`the advent of molecular gene transfer techniques and increased knowledge of the basic pathways
`of immune activation, the field of cancer immunotherapy has finally begun to develop novel
`and effective approaches for harnessing the immune system as a therapeutic agent. Current
`immunotherapy and gene therapy strategies, including methods of cytokine delivery and tumor-
`cell-based vaccines, are presented.
`
`Key words
`
`cytokine, gene therapy, immunotherapy, renal cell carcinoma, tumor vaccine.
`
`Introduction
`
`For at least 100 years, immunologists have proposed
`activating the immune system to specifically target and
`eradicate autologous tumor cells. The idea that tumor
`cells can be recognized as foreign to the host’s immune
`system is an essential component of tumor immunol-
`ogy. This concept of tumor cell recognition as foreign
`by their host was first postulated by Paul Ehrlich at the
`turn of the century. In 1943, Gross noted that when
`tumor cells were injected subcutaneously into synge-
`neic mice, the cells formed nodules that grew for a few
`days and then regressed.1 When tumor cells were re-
`injected into the mice, they failed to produce nodules
`or grow. This was interpreted to mean that the tumor
`cells did not grow because the mice had become
`immunologically resistant to the tumor, documenting
`the existence of tumor-associated antigens. In 1954,
`Billingham introduced the term ‘adoptive immunity’ to
`describe the acquisition of immunity as a result of the
`transference of immunologically competent cells rather
`
`Correspondence: Allan J Pantuck MD, University of
`California School of Medicine, Department of Urology,
`10833 Le Conte Avenue, Room 66-118 CHS, Los Angeles,
`USA. Email: apantuck@mednet.ucla.edu
`Presented at the 4th Urological Research Forum, 28–30
`January 2000, Okinawa, Japan.
`
`than of preformed antibody.2 In 1957, Prehn and Main
`demonstrated, further, that immunization of syngeneic
`mice with a given tumor protected the mice against a
`second challenge with the same tumor, but did not pro-
`tect them from other tumors.3 In 1959, Thomas sug-
`gested that the immune response might be able to rid
`the body of abnormal cells.4 His theories were later
`refined into the 1970 immune surveillance hypothesis
`of Burnet, which suggests that the immune system
`could recognize malignant cells as foreign and gener-
`ate a response against them and that only tumors capa-
`ble of evading the body’s surveillance would be able to
`grow.5 In 1972, Borberg successfully documented the
`ability of adoptively transferred immune cells to cause
`regression of established syngeneic tumors.6 Finally,
`the first successful clinical application of cellular ther-
`apy in humans was performed at the National Cancer
`Institute, Bethesda, USA, by Rosenberg in patients
`with metastatic melanoma and renal cell carcinoma.7
`Peripheral blood lymphocytes of patients were acti-
`vated ex vivo with IL-2 to generate lymphokine-
`activated killer cells (LAK) which, when reinfused into
`the patients, were capable of non-major histocompati-
`bility complex (MHC) restricted tumor lysis.
`With the advent of molecular gene transfer tech-
`niques and increased knowledge of the regulation of
`the immune response, effective methods for harnessing
`the immune system as a therapeutic agent are finally
`
`NOVARTIS EXHIBIT 2101
`Breckenridge v. Novartis, IPR 2017-01592
`Page 1 of 4
`
`

`

`S2 AJ Pantuck et al.
`
`being realized. The existence of an immune response
`against a tumor is based on changes in the surface
`components of the tumor cell that do not occur in its
`non-malignant counterpart and that give rise to struc-
`tures that may be antigenic. Tumor-associated antigens
`on the surfaces of malignant cells may be unique to the
`cancerous cells and absent from their normal counter-
`part, or tumor antigens may be present on normal cells
`but become unmasked on the malignant cell or re-
`present products that are present during embryonic
`development but are absent in the normal adult
`tissue. Immune effector mechanisms capable of de-
`stroying tumor cells in vivo include humoral (antibody-
`mediated) and cell-mediated cytotoxicity.
`These recent achievements in basic science have not
`passed over the clinical realm of genitourinary oncol-
`ogy. The use of intravesical immunotherapy for super-
`ficial bladder tumors was the first immune-based
`therapy for bladder cancer and has become the gold
`standard. Developments in immunotherapy have re-
`sulted in an improved outlook for patients presenting
`with advanced renal cell carcinoma as well as for those
`who develop both distant and local recurrences after
`failed curative treatment. These achievements repre-
`sent only the beginning of new directions to be pur-
`sued. The future prospects of cancer therapy will be,
`without doubt, built upon the foundation of current
`investigative efforts in gene and immune therapy.
`
`Immune and gene therapy for
`renal cell carcinoma
`
`Traditionally, there have been no other effective treat-
`ments for renal cell carcinoma (RCC) aside from
`surgery as RCC is radiation- and chemo-resistant.
`Metastatic RCC has a poor prognosis, with an average
`survival of only 6–12 months from the time of diagno-
`sis and with only a 6% objective response rate with
`conventional chemotherapy. Thus, additional therapy,
`mainly for patients with advanced RCC, is urgently
`needed. The first advance in this direction occurred
`when IL-2 was isolated and identified. The molecular
`cloning of IL-2 revolutionized the field of cancer
`immunotherapy and significantly altered the treatment
`of metastatic RCC.8 With the advent of recombinant
`DNA technology, the ability to produce large quantities
`of IL-2 has resulted in its widespread use. In a rela-
`tively short period of time, this agent has become
`a Federal Drug Administration approved treatment for
`metastatic RCC. Since then, other immunostimulatory
`cytokines have been identified and purified. To date,
`most studies investigating the use of cytokines are
`
`focusing on IFN-a, IL-2, combinations of these
`cytokines, or adoptive immunotherapy with tumor-
`infiltrating lymphocytes (TIL) or LAK cells. The role
`of other cytokines (IL-4, IL-7, IL-12 and GM-CSF) is
`currently under investigation.
`Impressive advances have occurred in the past two
`decades in the application of immunotherapy to treat
`RCC. At the University of California Los Angeles
`(UCLA), we have seen a progressive increase in
`response to treatment as therapy has evolved from
`systemic IFN-a administration (16%), to combination
`IFN + IL-2 (25%), to the current method of bulk TIL
`(33%) and CD8/TIL (40%). Patient characteristics that
`predict improved responsiveness to therapy have been
`identified and treatment protocols that decrease toxic-
`ity have been developed. The most encouraging results
`have been the improved rates of complete clinical
`response, most of which are durable and long-lasting.
`Further refinements in the treatment of renal cell
`disease with biologic and immunotherapeutic agents
`are still needed, yet there is no doubt that current
`immunotherapeutic protocols produce changes in the
`natural history of this disease and cause significant and
`lasting remissions in selected patients. Progress in
`understanding the genetic changes that are associated
`with the development of RCC has been made over sev-
`eral years. Thus, gene therapy for RCC has advanced
`further than in any other urological organ system.
`Intense efforts and progress have proceeded along sev-
`eral therapeutic paths.
`
`Immunotherapy
`
`It is well established that IL-2 has a beneficial activity
`in patients with advanced RCC; thus, a tumor vaccine
`seems appealing. By using a tumor vaccine, elevated
`cytokine concentrations can be achieved within the
`tumor causing an increase in MHC expression. By
`increasing the surface MHC expression, especially
`of HLA-Cw7, an immune response is anticipated
`once the TIL are able to recognize the MHC-restricted
`tumor-related peptides. By achieving locally high
`cytokine concentrations, the systemic toxicity that
`limits
`the efficacy of
`immunotherapy should be
`avoided.
`Initial studies with tumor vaccines in animal models
`have shown that the transfer of cytokine genes to
`tumor cells is feasible and can induce host antitumor
`effects.9 IL-2-transfected RCC cells inhibit the growth
`of parental tumor cells in rats.9 It has also been shown
`that the production of IL-2 is more intense after intra-
`tumor injection than after systemic administration of
`the transfecting agent. A synergistic antitumoral effect
`
`NOVARTIS EXHIBIT 2101
`Breckenridge v. Novartis, IPR 2017-01592
`Page 2 of 4
`
`

`

`was reported for retroviral transduction of the IL-2
`gene conjugated with systemic administration of IFN-
`a. This suggests that a synergistic, immunogenic effect
`can be obtained by using both local gene therapy and
`systemic, immune stimulation. IL-4, GM-CSF, HLA-
`137 and IFN-a gene transfection are other immune
`system modulators that may have a role in future
`tumor vaccines for RCC.10
`Recent phase I trials using tumor vaccines have
`been initiated in humans with metastatic RCC. Patients
`were given irradiated autologous tumor cells trans-
`fected in vitro with a retroviral vector carrying the
`GM-CSF gene. No significant toxicity was reported.
`One out of 16 patients had a partial response.
`Additionally, studies using genetically modified den-
`dritic cells and studies using the injection of cytokines
`into the tumor have been performed at UCLA, by
`using the HLA-B7 and IL-2 genes carried in a liposo-
`mal vector. In addition to these, at least three other
`tumor vaccine programs have been initiated using
`either intratumoral HLA-B7 or IL-2 gene transfection
`to enhance the immunogenicity of the tumor. At this
`time, although tumor vaccine-based gene therapy
`appears to be safe, its efficacy in metastatic RCC has
`yet to be proved.
`
`Gene therapy for RCC S3
`
`product in an attempt to reverse the cancer phenotype.
`Normal (wild-type) VHL gene was transfected into
`RCC cell lines lacking the normal expression of the
`gene. The wild-type VHL gene was attached to a con-
`stitutively activated cytomegalovirus promoter and put
`into a liposome vehicle. Transfection of the wild-type
`VHL gene had no effects on the transfected cell line
`growth in vitro, but the expression of the wild-type
`VHL gene resulted in growth suppression of other
`RCC cell lines. This study showed that the suppression
`of cell growth was specific to RCC cell lines, which
`implied that the VHL protein is important in control-
`ling the proliferation of kidney cells. Thus, gene
`replacement therapy using the wild-type VHL gene
`may have a role in treating patients with RCC,
`although the safety and efficacy of this treatment is yet
`to be defined.
`In vitro attempts to replace 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 meta-
`static lung lesions.13 The use of the p53 wild-type gene
`by intratumoral injection may prove to be efficacious
`in the future.
`
`Corrective gene therapy
`
`Cyto-reductive therapy
`
`From studies of familial RCC in patients with the von
`Hippel–Lindau syndrome, the molecular basis for
`tumorigenesis of the kidney is becoming clearer; loss
`of chromosome 3p in many sporadic and familial
`renal cell cancers has been noted,11 with restriction
`fragment length polymorphisms (RFLP). The von
`Hippel–Lindau (VHL) gene was identified at 3p25.5 of
`chromosome 3.11 It has been hypothesized that the
`VHL protein functions as a cell-cycle regulator, con-
`trolling cellular proliferation by restricting gene tran-
`scription, translation or repair. However, only 45–60%
`of all patients with sporadic RCC have a detectable
`mutation in the VHL gene. Furthermore, the pheno-
`typic expression of the VHL gene defect varies, with
`loss of the VHL gene product not always resulting in
`RCC.12 Thus, the defect in the VHL gene is probably
`influenced by many other yet to be defined epigenetic
`phenomena. Moreover, aberrations at chromosome 5,
`7, 14 and at the Y-chromosome have also been associ-
`ated with RCC. These factors may be able to act
`independently from the VHL locus, resulting in the
`development of RCC.
`Despite the limitations of the VHL gene as a target
`for gene therapy, initial studies have been performed in
`an attempt to replace the defective tumor suppressor
`
`A tumor marker has been recently identified for RCC.
`This new tumor antigen has been named G250. The
`function of this protein is unclear. High levels of G250
`antigen can be detected in up to 90% of all kidney can-
`cer cells, with normal renal parenchyma showing no
`detectable G250 antigen. This antigen has been used as
`a target for monoclonal antibody immunohistochemi-
`cal staining for diagnostic purposes and has been also
`used in radionuclide scans to localize tumor sites.14
`Because this antigen is found in a high proportion of
`RCC cases, it has the potential to be a target for gene
`therapy. Initial studies looking at cytokine-stimulated
`human RCC xenografts showed that the antitumor
`activity of the immune system could be enhanced by
`the administration of antibodies to G250. For now, it is
`unknown whether the G250 protein itself can stimulate
`an immune response. Furthermore, because expression
`of this gene product is also seen in normal bile ducts
`and normal gastric mucosa, the safety of an anti-G250
`treatment needs to be further tested.
`Thus, at this time, there is no perfect target for all
`patients who have RCC, although restoration of the
`wild-type VHL gene product and the development of
`targeted therapy against the G250 protein do hold sig-
`nificant promise for future trials.
`
`NOVARTIS EXHIBIT 2101
`Breckenridge v. Novartis, IPR 2017-01592
`Page 3 of 4
`
`

`

`S4 AJ Pantuck et al.
`
`Conclusion
`
`Although surgical management continues to be an
`effective
`treatment
`for organ-confined neoplastic
`disease, the treatment options for patients with dis-
`seminated cancer are limited. Immunotherapy has
`demonstrated significant success in the management of
`advanced RCC. Recent exciting research has demon-
`strated that prostate cancer may also be susceptible to
`immuno-therapeutic protocols, finally offering a glim-
`mer of hope to physicians treating this very prevalent
`and morbid disease. Molecular-based therapy has
`many potential applications
`in
`the
`treatment of
`advanced genitourinary cancers. The reinsertion of
`inactivated tumor suppressor genes, the inactivation
`of oncogenes, the insertion of immunomodulatory
`genes and the insertion of suicide genes have all been
`used to treat genito-urinary malignancies, in vitro and
`in animal models. Progress is being made in better
`understanding the genetic and cellular mechanisms
`that underlie tumorigenesis. Human clinical trials are
`already in phase I testing in some tumor systems,
`including RCC,
`transitional cell carcinoma and
`prostate cancer. However, limitations still have to be
`overcome. Safe and effective gene vectors will be
`needed to carry the therapeutic gene to the host cell.
`Treatments need to be tailored so that the desired
`effects occur only in the tumor cells. In conclusion,
`molecular-based therapy is appealing because of its
`ability to treat cancer at the level of the gene defect
`that causes the malignant phenotype, and it offers
`novel and exciting approaches for the treatment and
`ultimate eradication of cancer.
`
`References
`
`1 Gross L. Intradermal immunization of C3H mice
`against a sarcoma that originated in an animal of the
`same line. Cancer Res. 1943; 3: 326–33.
`
`2 Billingham RE, Brent L, Medawar PB. Quantitative
`studies on tissue transplantation immunity. Volume II:
`The origin, strength and duration of activity and adop-
`tively acquired immunity. Proc. R. Soc. Biol. 1954;
`143: 58–80.
`3 Prehn RT, Main JM. Immunity to methylcholanthrene-
`induced sarcomas. J. Natl Cancer Inst. 1957; 18:
`769–78.
`4 Thomas L, Lawrence HC. Cellular and Humoral
`Aspects of the Hypersensitive States. Hoeber-Harper,
`New York, 1959.
`5 Burnet FM. The concept of immunological surveil-
`lance. Prog. Exp. Tumor Res. 1970; 13: 1–27.
`6 Borberg H, Oettgen HF, Choudry K et al. Inhibition of
`established transplants of chemically induced sarcomas
`in syngeneic mice by lymphocytes from immunized
`donors. Int. J. Cancer 1972; 10: 539–47.
`7 Rosenberg SA, Lotze MT, Muul LM et al. Observa-
`tions on the systemic administration of autologous
`lymphokine-activated killer cells and recombinant
`interleukin-2 in patients with metastatic cancer. N.
`Engl. J. Med. 1985; 313: 1485–92.
`8 Smith KA. Interleukin-2: Inception, impact, and impli-
`cations. Science 1988; 240: 1169–76.
`9 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: 207–16.
`10 Gitlitz BJ, Belldegrun A, Figlin RA. Immunotherapy
`and gene therapy. Semin. Urol. Oncol. 1996; 14:
`237–43.
`11 Glenn GM, Linehan WM, Hosoe S et al. Screening for
`von Hippel–Lindau disease by DNA polymorphism
`analysis. JAMA 1992; 267: 1226–31.
`12 Latif F, Tory K, Gnarra J et al. Identification of the von
`Hippel–Lindau disease tumor suppressor gene. Science
`1993; 260: 1317–20.
`13 Moon WC, Kim YS, Moon CS. Experimental p53 gene
`therapy in renal cell carcinoma. J. Urol. 1996; 155:
`653A.
`14 Oosterwijk E, Bander NH, Divgi CR et al. Antibody
`localization in human renal cell carcinoma: A phase I
`study of monoclonal antibody G250. J. Clin. Oncol.
`1993; 11: 738–50.
`
`NOVARTIS EXHIBIT 2101
`Breckenridge v. Novartis, IPR 2017-01592
`Page 4 of 4
`
`

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