SlideShare ist ein Scribd-Unternehmen logo
1 von 52
Downloaden Sie, um offline zu lesen
Kidney Cancer:
Recent Advances in Diagnosis
and Management
                                                           Saturday, May 17, 2008
                                                               6:00- 8:00 p.m.




               COURSE 08 EC

                           FACULTY

    W. Marston Linehan, M.D., Ph.D.
            Course Director

                   Peter A. Pinto, M.D.
                 Jeffrey A. Sosman, M.D.




                American Urological Association
                  Education and Research Inc.
               2008 Annual Meeting, Orlando, FL
                       May 17-22, 2008

   Sponsored by: The American Urological Association Education and Research, Inc.
Meeting Disclaimer
Regarding materials and information received, written or otherwise, during the 2008 American Urological
Association Education and Research, Inc. Annual Meeting Instructional/Postgraduate MC/EC and Dry Lab
Courses sponsored by the Office of Education:

The scientific views, statements, and recommendations expressed in the written materials and
during the meeting represent those of the authors and speakers and do not necessarily represent
the views of the American Urological Association Education and Research, Inc.®

Reproduction Permission

Reproduction of all Instructional/Postgraduate, MC/EC and Dry Lab Courses is prohibited without written
permission from individual authors and the American Urological Association Education and Research, Inc.

These materials have been written and produced as a supplement to continuing medical education activities
pursued during the Instructional/Postgraduate, MC/EC and Dry Lab Courses and are intended for use in that
context only. Use of this material as an educational tool or singular resource/authority on the subject/s
outside the context of the meeting is not intended.

Accreditation

The American Urological Association Education and Research, Inc. is accredited by the Accreditation
Council for Continuing Medical Education (ACCME) to provide continuing medical education (CME) for
physicians. The American Urological Association Education and Research, Inc. takes responsibility for the
content, quality, and scientific integrity of the CME activity.

CME Credit

The American Urological Association Education and Research, Inc. designates each Instructional Course
educational activity for a maximum of 1.5 AMA PRA Category 1 credits™; each Postgraduate Course for a
maximum of 3.25 AMA PRA Category 1 credits™; each MC Course for a maximum of 1.0 AMA PRA
Category 1 credits™; each EC Course for a maximum of 2.0 AMA PRA Category 1 credits™; each MC Plus
Course for a maximum of 2.0 AMA PRA Category 1 credits™; and each Dry Lab Course for a maximum of
2.5 AMA PRA Category 1 credits™. Physicians should only claim credits commensurate with the extent of
their participation in the activity.

Disclosure Policy Statement

As a provider accredited by the Accreditation Council for Continuing Medical Education (ACCME), the
American Urological Association Education and Research, Inc., must insure balance, independence,
objectivity and scientific rigor in all its sponsored activities.

All faculty participating in an educational activity provided by the American Urological Association Education
and Research, Inc. are required to disclose to the audience any relevant financial relationships with any
commercial interest to the provider. The intent of this disclosure is not to prevent a faculty with relevant
financial relationships from serving as faculty, but rather to provide members of the audience with
information on which they can make their own judgments. The American Urological Association Education
and Research, Inc. must resolve any conflicts of interest prior to the commencement of the educational
activity. It remains for the audience to determine if the faculty’s relationships may influence the educational
content with regard to exposition or conclusion. When unlabeled or unapproved uses are discussed, these
are also indicated.

                                          Evidence-based Content

As a provider of continuing medical education accredited by the Accreditation Council for Continuing Medical
Education (ACCME), it is the policy of the American Urological Association Education and Research, Inc. to
review and certify that the content contained in this CME activity is evidence-based, valid, fair and balanced,
scientifically rigorous, and free of commercial bias.
2008 AUA Annual Meeting


08 EC Kidney Cancer: Recent Advances in Diagnosis and Management

5/17/2008                                                                                    6:00 - 8:00 p.m.


Disclosures

According to the American Urological Association’s Disclosure Policy, speakers involved in continuing
medical education activities are required to report all relevant financial relationships with any commercial
interest to the provider by completing an AUA Disclosure Form. All information from this form is provided to
meeting participants so that they may make their own judgments about a speaker’s presentation. Well in
advance of the CME activity, all disclosure information is reviewed by a peer group for identification of
conflicts of interest, which are resolved in a variety of ways.

The American Urological Association does not view the existence of relevant financial relationships as
necessarily implying bias, conflict of interest, or decreasing the value of the presentation.

Each faculty member presenting lectures in the Annual Meeting Instructional or Postgraduate, MC or EC
and Dry Lab Courses has submitted a copy of his or her Disclosure online to the AUA. These copies are on
file in the AUA Office of Education.

This course has been planned to be well balanced, objective, and scientifically rigorous. Information and
opinions offered by the speakers represent their viewpoints. Conclusions drawn by the audience members
should be derived from careful consideration of all available scientific information.

The following faculty members(s) declare a relationship with the commercial interests as listed below,
related directly or indirectly to this CME activity. Participants may form their own judgments about the
presentations in light of full disclosure of the facts.


Faculty Disclosure
W. Marston Linehan, M.D., Ph.D.
Course Director
National Cancer Institute: Meeting Participant or Lecturer

Peter A. Pinto, M.D.
Nothing to disclose

Jeffrey A. Sosman, M.D.
Nothing to disclose
Disclosure of Off-Label Uses

The audience is advised that this continuing medical education activity may contain reference(s) to
unlabeled or unapproved uses of drugs or devices. Please consult the prescribing information for full
disclosure of approved uses. Faculty and speakers are required to disclose unlabeled or unapproved
use of drugs or devices before their presentation or discussion during this activity.




A special AUA value for your patients:
www.UrologyHealth.org is a joint AUA/AFUD patient education web site that provides accurate
and unbiased information on urologic disease and conditions. It also provides information for
patients and others wishing to locate urologists in their local areas. This site does not provide
medical advice. The content and illustrations are for informational purposes only. This
information is not intended to substitute for a consultation with a urologist. It is offered to educate
the patient, and their families, in order for them to get the most out of office visits and
consultations.
Commercial Support Acknowledgements

AUA Office of Education thanks the companies and representatives who support
continuing education of physicians with unrestricted educational grants, displays
at educational meetings, and their expertise. This is a valuable resource to
everyone.

The AUA Office of Education salutes the following company for providing an
educational grant in support of the Kidney Cancer: Recent Advances in
Diagnosis and Management course at the 2008 Annual Meeting.




                       Wyeth Pharmaceuticals


The scientific programs and faculty are independently developed and finalized by the Chair of
Education, the Director of Education and Scientific Program Director with no influence or input
from these or any companies providing either an unrestricted educational grant or technical
support.

        Comments or letters regarding this program and/or company involvement in an
        educational meeting may be addressed to:

        Jan Baum, M.A.
        Director of Education
        American Urological Association
        1000 Corporate Boulevard
        Linthicum, MD 21090
        (410) 689-3930
Recent Advances in Diagnosis and Management
                of Kidney Cancer
                W. Marston Linehan, M.D.
             Chief, Urologic Oncology Branch
                 National Cancer Institute
                    Bldg 10 Rm 1-5942
             Bethesda, Maryland 20892-1107
                    Tel: (301) 496-6353
                    Fax: (301) 402-0922
                           Email:
                  Email: WML@nih.gov

                      Peter Pinto, M.D.
       Senior Investigator, Urologic Oncology Branch
                  National Cancer Institute
                     Bldg 10 Rm 2-5940
              Bethesda, Maryland 20892-1107
                     Tel: (301) 496-6353
                    Fax: (301) 402-0922
                Email : pintop@mail.nih.gov

                  Jeffrey A. Sosman, M.D.
       Professor of Medicine (Hematology/Oncology)
                 Vanderbilt-Ingram Cancer
                    777 Preston Building
                 Nashville, TN 27232-6307
                   Office (615) 343-7602
            Email: jeff.sosman@Vanderbilt.Edu
RECENT ADVANCES IN DIAGNOSES AND MANAGEMENT
                            OF KIDNEY CANCER
                           Saturday, May 17, 2008
                              6:00PM - 8:00PM



From     To         Speaker                        Topic


6:00 PM - 6:20 PM   W. Marston Linehan, M.D.       I.  Molecular Genetics of Renal Carcinoma:
                                                       Implications for the Urologic Surgeon
                                                   II. Hereditary Forms of Renal Cancers

6:20 PM - 7:00 PM   Peter A. Pinto, M.D.           III. Techniques and Indications
                                                        A) Laparoscopic Nephrectomy
                                                        B) Laparoscopic Partial Nephrectomy
                                                   IV. Robotic Assisted Nephrectomy and
                                                        Partial Nephrectomy

7:00 PM - 7:40 PM   Jeffrey A. Sosman, M.D.        V. Role of Targeted Systemic Therapy
                                                       In the Management of Advanced Clear Cell
                                                       And Non-Clear Cell Kidney Cancer
                                                      A) Sorafenib
                                                      B) Sunitinib
                                                      C) Bevacizumab
                                                      D) Temsirolimus

7:40 PM - 8:00 PM   W. Marston Linehan, M.D.       VI. A) Management of Patients Who Present with
                    Peter A. Pinto, M.D.                  Advanced Disease with Kidney In Place
                    Jeffrey A. Sosman, M.D.            B) Role of Adjuvant Therapy in the
                                                          Management of Patients with High Risk
                                                          Kidney Cancer




                                               2
I. Introduction
        Kidney cancer affects over 39,000 Americans annually and it is estimated that nearly
13,000 will die of this disease in the United States each year. Patients who are found to have the
disease when it is localized to the kidney can have up to a 95% 5 and 10 year survival rate.
Patients who present to their urologic surgeon with locally advanced disease have up to a 65%
chance of surviving 5 years. Those who present with advanced disease have an 18% 2 year
survival rate.1,2
        The advent of laparoscopic surgery has provided new and significantly less morbid
surgical strategies for the management of patients with localized, locally advanced and even
advanced renal carcinoma. There have been exciting advances in the development of new
minimally invasive strategies for the management of patients with localized kidney cancer
involving radio frequency ablation therapy as well as cryotherapy. Recent advances in
immunologic and molecular targeted forms of therapy have provided new hope for patients with
advanced renal cell carcinoma.3,4

       1. Molecular Genetics of Renal Carcinoma: Implications for the
       Urologic Surgeon (W. Marston Linehan, M.D.)
       Kidney cancer is not a single disease, it is made up of a number of different types of
cancer that occur in the kidney; each with a different histology, having a different clinical course,
responding differently to therapy and caused by different genes.




       Figure 1 Kidney cancer is made up of a number of different types of cancer, each with a
       different histology, with a different clinical course, responding differently to therapy and
       caused by different genes. From Linehan, et al.5

         This section of the course will update you on current studies of the molecular genetics of
sporadic and familial renal cancer, background on kidney cancer tumor suppressor genes and
oncogenes, data for why we conclude that the von Hippel Lindau disease gene and the Met genes
are critical genes for both hereditary and non-familial, sporadic kidney cancers, for how these
genes fit the definitions of cancer genes and how these types of findings could lead to better
methods for early diagnosis, prevention and, potentially, therapy of renal cancers will be
presented.



                                                      3
2. Tumor Suppressor Genes and Oncogenes
                A. Tumor Suppressor Genes
        The pioneering studies of Dr. Alfred Knudson led to the development of a hypothesis that
the most basic aspect of cancer would involve what is called a tumor suppressor gene. Instead of
cancer being the result of activation of a gene such as an oncogene, which drives the abnormal
proliferation that we know of as cancer, Knudson hypothesized that, instead, cancer could be the
result of inactivation of a gene, a so called tumor suppressor gene, whose normal function was to
regulate or control cellular growth.
        We each have two copies of each gene, one maternal and the other paternal. If both
copies of a tumor suppressor gene were inactivated (Knudson “two-hit” hypothesis) by either
loss of DNA (DNA sequence deletion), or another mechanism such as mutation, transformation
would result.
        Knudson hypothesized that if this were the case in a sporadic, non-hereditary cancer, and
there were a hereditary variant of the same malignancy, that the same gene might be involved in
the origin of both. This is the case in retinoblastoma, where both copies of the retinoblastoma
gene are inactivated in both the sporadic and hereditary form of the disease. Abnormalities of
tumor suppressor genes have been found in a number of solid human tumors, including breast,
bladder and, as we shall see, kidney cancer. The VHL kidney cancer tumor suppressor gene,
the gene for clear cell renal carcinoma, is a classic tumor suppressor gene located on
chromosome 3.
                B. Oncogene
        An oncogene is a “gain of function gene”; i.e., a gene in which a mutation of the gene
results in activation of the gene, resulting in the unregulated growth that we know of as cancer.
Often an oncogene may be “duplicated” in a cancer, resulting in three copies of the gene (instead
of two copies). This is called trisomy. The c-Met oncogene, the gene for Hereditary
Papillary Renal Carcinoma (HPRC), is a classic oncogene located on chromosome 7.




                                               4
II. Hereditary Forms of Renal Cancers
         Kidney cancer is like retinoblastoma and colon cancer in that there is both a sporadic and
a familial form. Familial kidney cancer is far more prevalent than had been previously imagined.
It is estimated that 5% of kidney cancer is familial (hereditary). There are four types of
hereditary kidney cancer. The well described is clear cell renal cancer associated with von
Hippel Lindau (VHL); there is a hereditary form of type 1 papillary renal carcinoma (HPRC); the
third type is hereditary chromophobe renal carcinoma associated with Birt Hogg Dubé (BHD)
and the fourth is a hereditary type of papillary type 2 renal carcinoma associated with Hereditary
Leiomyomatosis Renal Carcinoma (HLRCC).


                       Table 1 Inherited Forms of Renal Carcinoma


                     1. Von Hippel Lindau (VHL)
                            Clear Cell RCC
                     2. Hereditary Papillary Renal Carcinoma (HPRC)
                            Papillary Type 1 RCC
                     3. Birt Hogg Dubé (BHD)
                            Chromophobe RCC/Oncocytoma
                     4. Hereditary Leiomyomatosis RCC (HLRCC)
                            Papillary Type 2 RCC


        These familial forms of kidney cancer are different from sporadic, non-familial kidney
cancer in that they tend to be multifocal, bilateral and occur at a younger age, suggesting a genetic
predisposition to develop these cancers.

       1. Von Hippel Lindau (VHL)
                A. Renal Carcinoma Gene Localization Studies: Inherited Renal Cancer
       To identify the disease gene for clear cell kidney cancer the familial form of renal cell
carcinoma associated with von Hippel Lindau (VHL) was studied. Von Hippel Lindau is an
autosomal dominant hereditary cancer syndrome in which affected individuals are at risk to develop
tumors in a number of organs, including the kidneys.
       VHL patients are at risk to develop multiple, bilateral renal carcinomas and cysts, cerebellar
and spinal hemangioblastomas, pheochromocytomas, retinal angiomas, epididymal cystadenomas,
pancreatic cysts and islet cell tumors and tumors in the inner ear, endolymphatic sac tumors.




                                                 5
Table 2 Clinical Features of VHL


                        h   Kidney tumors (clear cell RCC)
                        h   Adrenal tumors (pheochromocytoma)
                        h   Pancreatic tumors (islet tumors)
                        h   CNS tumors (hemangioblastoma)
                        h   Retinal tumors (angioma)
                        h   Endolymphatic sac tumors (inner ear)
                        h   Epididymal cystadenoma


                 B. VHL: Clinical Evaluation
        Clinical evaluation of VHL patients involves MRI of the brain and spine (for CNS
hemangioblastomas), abdominal CT and ultrasound (for evaluation of renal, adrenal and
pancreatic manifestations of VHL), ophthalmologic evaluation (for evaluation for retinal
hemangiomas), audiometric and ENT evaluation (to search for presence of ELST tumors),
testicular ultrasound (to evaluate epididymal cystadenomas).


                        Table 3 Clinical Evaluation-VHL


                    •       VHL gene germline mutation testing
                    •       MRI of the brain and spine
                    •       Abdominal CT and ultrasound
                    •       Ophthalmologic evaluation
                    •       Audiometric and ENT evaluation
                    •       Testicular Ultrasound
                    •       Metabolic evaluation (catechols)

                C. VHL: Surgical Management
        Surgical management of the renal manifestations of VHL patients involves nephron
sparing surgery whenever possible. Patients with small renal tumors, generally under 2.5 cm, are
often managed with expectant management. When the tumors reach 3 cm, surgery is often
recommended. As it has been estimated that there can be up to 600 tumors per kidney in VHL
patients, surgical resection of renal lesions is not considered “curative.” Rather, it is considered
that surgical management will hopefully “set back the clock”; i.e. help prevent metastasis.
Historically 35 to 45% of VHL patients have died of complications of metastatic renal cell
carcinoma. The decision to recommend surgery must balance the risk of metastasis with the
morbidity of surgery. This is often a complex issue in VHL patients with multisystem
manifestations. When surgery is performed, thorough evaluation of the kidney with
intraoperative ultrasound is considered a valuable adjunct to the surgical procedure. This allows


                                                 6
the surgeon to localize renal tumors and cysts and to perform as thorough and safe a procedure as
possible.




        Renal Lesions in VHL Patient                  Microscopic VHL RCC Focus

It is important that VHL patients undergo a complete screening and metabolic evaluation prior to
a surgical procedure, to rule out such unsuspected manifestations as a CNS hemangioblastoma or
pheochromocytoma.

                D. The VHL gene is on Chromosome 3
       In order to identify the gene associated with VHL we evaluated affected and at risk
individuals for VHL. DNA was extracted from blood of over 4,000 individuals. 450 patients
were screened at the NIH for the presence of von Hippel Lindau. In order to localize this kidney
cancer disease gene genetic linkage analysis was utilized, which narrowed the area of the VHL
gene to a small region on chromosome 3, which is the same region where we showed
abnormalities in tumor tissue from patients with non-familial, clear cell kidney cancer.




             Mutations of the VHL Gene in the Germline of VHL Patients From Chen et al.6


              E. The VHL gene is the gene for sporadic, non-inherited clear cell RCC




              Mutations of the VHL gene is tumors from patients with sporadic, non-
              hereditary clear cell kidney cancer. From Gnarra, et al. 7


       VHL mutations have been identified in a high percentage of tumors from patients with
sporadic, clear cell renal carcinomas. VHL somatic mutations have been detected in the earliest,


                                                  7
localized clear cell tumors. No mutations have been found in papillary, chromophobe, collecting
duct or other types of renal tumors.

                F. Molecular Targeting the VHL gene in clear cell kidney cancer
        It is hoped that understanding the pathways for the genes that cause kidney cancer will
lead to the development of more effective forms of therapy for patients with this disease. The
VHL clear cell kidney cancer gene was identified in 1993 and since that time an intensive effort
has been underway to identify how damage to this gene leads to kidney cancer. The VHL gene
product has been found to be important regulator of Hypoxia Inducible Factor (HIF). HIF is a
transcription factor which regulates a number of downstream genes known to be important in
cancer, such as vascular endothelial growth factor (VEGF), platelet derived growth factor
(PDGF), erythropoietin, etc. Scientists are currently evaluating the role of small molecules that
block this pathway as potential strategies for therapy.




       VHL Kidney Cancer Gene Pathway                        RCC Molecular targeting: anti-VEGF antibody.
       Adapted from Linehan, et al.5
The FDA recently approved three new agents, sunitinib, sorafenib and temsirolimus for the
treatment of patients with advanced kidney cancer.

                             Targeting VHL/HIF in Clear Cell RCC

                                                    VHL



                                                    HIF




                                     VEGF           PDGF        TGF-α

                                     VEGFR         PDGFR        EGFR



                                             Sunitinib




       Sunitinib The partial response rate in patients with advanced renal carcinoma
       treated with sunitinib has been reported to be 31% and there was an increase in
       progression free survival in patients treated with sunitinib versus interferon.8 The
       most commonly reported side effects with sunitinib involve diarrhea, skin
       discoloration, mouth irritation, weakness, and altered taste. Patients treated with
       sunitinib may also experience, high blood pressure, fatigue, bleeding, and
       swelling.


                                                         8
Targeting VHL/HIF in Clear Cell RCC

                                                       VHL



                                                           HIF




                                     VEGF              PDGF        TGF-α

                                    VEGFR             PDGFR       EGFR

                                     Raf                           Raf
                                               Sorafenib

                                   Sorafenib                     Sorafenib




       Sorafenib The partial response rate in patients with advanced renal carcinoma
       treated with sorafenib has been reported to be 10% and the progression free
       survival was 5.5 months versus 2.8 months for patients treated with placebo.9



                            Targeting VHL/HIF in Clear Cell RCC

                                                       VHL



                                                           HIF    mTOR



                                                                             Temsirolimus
                                     VEGF              PDGF        TGF-α

                                    VEGFR             PDGFR       EGFR




       Temsirolimus (inhibitor of mammalian target of rapamycin-MTOR)
       Temsirolimus, the MTOR-inhibiting agent, has been found to have activity in
       patients with advanced, poor prognosis kidney cancer. In a recent report of a
       randomized three-arm phase 3 trial, temsirolimus was found to have an overall
       statistically significant survival benefit versus treatment with interferon alone
       (10.9 versus 7.3 months). This study confirmed MTOR as a valid target in kidney
       cancer and that temsirolimus monotherapy prolongs survival in patients with poor
       prognosis kidney cancer. 10

               G. Adjuvant Treatment
The role of agents such as sunitinib and sorafenib in the adjuvant setting are not known. To
address this question, a randomized phase 3 trial involving 1332 patients, supported by the
Eastern Cooperative Oncology Group (ECOG), the Cancer Trials Support Unit CTSU), the
Southwest Oncology Group (SWOG), the Cancer and Leukemia Group B (CALGB) and the
National Cancer Institute of Canada (NCIC). In this trial, patients with T1B, T2 or T4 (N0-N2)
will be randomized to receive one year of sunitinib, sorafenib or placebo after nephrectomy.11
Information about the trial can be found at the Kidney Cancer Association web site.



                                                            9
2. Hereditary Papillary Renal Cell Carcinoma: HPRC
               A. HPRC: Clinical Features
       Hereditary Papillary Renal Carcinoma is a hereditary cancer syndrome in which affected
individuals are at risk for the development of bilateral, multifocal, type 1 papillary renal
carcinoma.12 HPRC patients are at risk for the development of up to 3,000 tumors per kidney,
which are uniformly of type 1 papillary histologic type.13




              Figure 2 Hereditary Papillary Renal Carcinoma (HPRC) is a hereditary cancer
              syndrome where affected individuals are at risk for the development of bilateral,
              multifocal, type 1 papillary renal carcinoma. From Linehan, et al.5



                     Table 4 Hereditary Papillary Renal Carcinoma
                     (HPRC)


                   h Clinical Features
                       h Bilateral, multifocal papillary renal cell carcinoma
                       h Type I papillary renal carcinoma


                   h Clinical Evaluation
                       –    C-Met gene germline mutation testing
                       –    Abdominal CT and ultrasound




                                                  10
B. Met is the Hereditary Papillary Renal Carcinoma gene
        The gene for type 1 papillary renal carcinoma associated with Hereditary Papillary Renal
Carcinoma is the Met proto-oncogene on chromosome 7.14,15 Met is a normal gene (a proto-
oncogene) that codes for Met, the cell surface receptor for the ligand hepatocyte growth factor.
Met becomes an oncogene when activating mutations occur in the tyrosine kinase domain of this
gene in the germline of HPRC patients. A simple blood test is available to determine if the at-
risk individual carries the germline Met mutation.




       Figure 3 Met, the cell surface receptor for the ligand, hepatocyte growth factor, is the gene
       for Hereditary Papillary Renal Carcinoma. Germline mutations of Met are found in
       affected individuals in HPRC kindreds. From Linehan, et al 5




                                                     11
3. Birt Hogg Dubé (BHD): Chromophobe Kidney Cancer
       Birt Hogg Dubé is a hereditary syndrome in which affected individuals are at risk for the
    development of kidney cancer, cutaneous lesions and pulmonary cysts.




              Figure 4 Birt Hogg Dubé (BHD) is a hereditary cancer syndrome in which patients
              are at-risk for the development of cutaneous lesions (fibrofolliculoma), pulmonary
              cysts and kidney cancer. From Linehan et al.5

   The kidney tumors can be chromophobe, hybrid/oncocytic, papillary or clear cell RCC.16 The
BHD gene was recently identified on chromosome 1717 and a blood test is now available for
determination of whether at-risk individuals are affected with BHD.18

                             Table 5 Clinical Features of Birt Hogg Dubé
                             (BHD)

                            h Cutaneous nodules (hair follicle tumors,
                              fibrofolliculoma) on the face and neck
                            h Pulmonary cysts
                            h Renal tumors
                               h   Chromophobe RCC
                               h   Oncocytic hybrid RCC
                               h   Clear cell RCC
                               h   Oncocytoma




                                                  12
Figure 5 BHD associated kidney cancer can be chromophobe, hybrid/oncocytic
RCC or oncocytoma. From Linehan, et al.5




        Table 6 Clinical Evaluation-BHD


    •     BHD gene germline mutation testing
    •     Dermatologic evaluation-skin biopsy
    •     Lung CT
    •     Abdominal CT/ultrasound




                                  13
4. Hereditary Leiomyomatosis Renal Cell Carcinoma (HLRCC)
               A. HLRCC: Clinical Features
       Hereditary leiomyomatosis renal cell carcinoma (HLRCC) is a hereditary cancer
       syndrome in which affected individuals are at risk for the development of cutaneous and
       uterine leiomyomas (fibroids) and an aggressive form of type 2 papillary kidney cancer.19




              Figure 6 HLRCC is a hereditary cancer syndrome in which affected individuals are
              at-risk for the development of cutaneous and uterine leiomyomas and type 2
              papillary renal carcinoma. From Linehan, et al.5




                Table 7 Clinical Features of Hereditary
                Leiomyomatosis Renal Cell Carcinoma (HLRCC)

                h    Cutaneous nodules (leiomyomas)
                h    Uterine leiomyoma (fibroids)
                h    Uterine leiomyosarcoma
                h    Renal tumor (type 2 papillary RCC)
                    h Often solitary
                    h Aggressive, early to metastasize


                B. Fumarate Hydratase (FH) is the gene for HLRCC
The Krebs cycle enzyme, fumarate hydratase (FH) is the gene for the hereditary form of Type II
papillary renal carcinoma associated with Hereditary Leiomyomatosis Renal Carcinoma
(HLRCC) and there is a blood test available to determine whether or not a patient is affected
with HLRCC.20 These tumors can be very aggressive21 and partial nephrectomy is often not
recommended for these patients. Early intervention with nephrectomy is recommended as these
aggressive tumors may spread early.




                                                14
Table 8 Clinical Evaluation-HLRCC


             •       Fumarate hydratase (FH) gene germline
                     mutation testing
             •       Dermatologic evaluation-skin biopsy
             •       Abdominal CT/ultrasound
             •       Pelvic CT/uterine ultrasound



       5. Familial Renal Carcinoma (FRC)
        Recently scientists in Iceland have performed studies suggesting that genetic
susceptibility may be a major component in the development of ordinary, “sporadic” renal
carcinoma. In their initial study, 68% of individuals in Iceland who had kidney cancer had up to
a second degree relative (a second cousin) with kidney cancer. This work suggests that it is
important to ask all patients with renal carcinoma whether any other family member also was
affected with renal carcinoma. Urologic surgeons at the NCI are currently studying families in
which multiple members are affected with kidney cancer (FRC) in order to identify the genetic
basis of this form of kidney cancer.22




                 Figure 7 Studies have shown that there may be a strong hereditary
                 component to “sporadic” kidney cancers. Investigators are currently
                 intensively studying families in which more than one family member has
                 kidney cancer in order to determine the gene(s) for familial renal
                 carcinoma. From Linehan, et al.5



                                                    15
III. Techniques and Indications (Peter Pinto, M.D.)

       A) Laparoscopic Nephrectomy
Surgical Technique: Transperitoneal Approach
        Positioning
        The following will describe the operative technique for a transperitoneal left laparoscopic
nephrectomy. The patient undergoes general endotracheal anesthesia, prophylactic antibiotics
and placement of an orogastric tube and urinary catheter. While supine the lower midline
incision is marked with an indelible pen. This is done prior to positioning to allow the incision
for possible specimen extraction to be symmetric in reference to the midline. This is difficult to
judge after the patient is positioned. The patient is then placed in a modified left flank position
with a bump under their left side. The right arm is tucked and the left arm is folded over
cushions across the chest. (Picture 1)




             Picture 1) Positioning for transperitoneal left laparoscopic renal surgery.
             From Pinto, et al.23

       All pressure points are checked and padded. The patient is secured to the table with 3
inch wide silk tape. In order to confirm that the patient is adequately secured, the table is
maximally tilted prior to draping.

        Port Placement
        The operating table is returned to a neutral position and the patient is prepped and draped
in the standard fashion. The surgeon and assistant stand on the patient’s right side and the scrub
nurse on the left side by the legs. In cases where a robotic camera holder is used, it is secured to
the table at the level of the patient’s right shoulder prior to draping. With the aid of a Veress
needle, pneumoperitoneum is established at a pressure of 20 mmHg. Four trocars are
subsequently placed: a 10mm periumbilical port for the camera, a 12mm port in the left
midaxillary line at the level of the umbilicus, a 12mm port in the midline of the premarked
pfannensteil incision, and a 5mm trocar in the midline, halfway between the umbilicus and


                                                    16
xyphoid. (Picture 2) In obese patients, the port placement is altered slightly. In these cases, the
ports are shifted laterally, with the camera port at the level of the umbilicus, but lateral to the
rectus muscle. (Figure 1)




            Picture 2) Port placement for left laparoscopic nephrectomy/partial nephrectomy.
                       Lower midline port can be extended for specimen retrieval.




       Figure 1) Port placement shifted laterally for obese patient. From Williams et al.24

        A visualizing trocar is used to place the first port, allowing the remaining three to be
placed under vision. Even in the setting of previous abdominal surgery, initial entry with a
veress and visualizing trocar has been shown to be safe. ( J Urol 170, 61–63, July 2003)
All trocars are secured to the skin with suture to avoid inadvertent removal during surgery.




                                                    17
Procedure
         The table is then tilted maximally to the right to help the bowel fall medially. This can be
facilitated by an assistant using a paddle retractor through the low midline port. The
retroperitoneal space is entered by incising the line of Toldt sharply from the spleen to the iliac
vessels. (Figure 2)




                         Figure 2) Entering the retroperitoneal space



         As the descending colon is reflected medially care must be taken to develop the plane
between the mesocolon and anterior gerota’s fascia. If not, a common mistake is to dissect
lateral to the kidney. This divides the renal attachments laterally and makes future hilar
dissection more difficult. In order to obtain proper exposure to the renal hilum and especially for
upper pole tumors, the splenophrenic ligament must be incised. Take care to avoid inadvertent
injury to the stomach and diaphragm as the spleen is being mobilized. Once completely
mobilized, the spleen, tail of pancreas and splenic flexure will fall medially out of the operative
field.
         Upper pole dissection is then carried out. The lienorenal ligaments are divided sharply in
order to avoid inadvertent splenic lacerations. This dissection is carried out toward the renal
hilum in order to separate the adrenal gland and expose the adrenal vein as it enter the renal vein.
In cases where the tumor location does not necessitate taking the adrenal gland, the renal vein
may be transected lateral, preserving the adrenal vein’s entry into the renal vein. Alternatively,
the adrenal vein can be clipped and divided. (Figure 3)




                                                  18
Figure 3) Upper pole mobilization when needed

With the upper pole of the kidney mobilized, attention is turned to the lower pole of the kidney,
ureter, gonadal and renal hilum. Dividing the colorenal ligaments allows the colon to fall
medially and exposes the lower pole, gonadal vessels and ureter. (Figure 4)




                       Figure 4) Lower pole, ureteral and gonadal dissection
         A plane is developed between the psoas muscle and the gonadal/ureteral packet. If the
ureter is difficult to identify, or obscured by a large lower pole mass, the dissection can be started
distally at the level of the iliac vessels and continued proximally to the renal vein. At the level of
the renal hilum lumbar vessels are ligated and divided as necessary. The neuro-lymphatic and
fibrofatty tissue surrounding the renal artery and vein is ligated and divided isolating the artery
down to its take off from the aorta. This dissection also allows for a hilar lymphadenectomy to be
performed if indicated. The renal artery and vein can be controlled with titanium or polymer
clips (leaving 3 on the patient side) or an endoscopic linear cutter stapler with a vascular staple
load (Figure 5).




                                                  19
Figure 5) Ligation of the renal vessels

       With the vessels divided, the ureter is ligated and the remaining lateral, posterior and
superior attachments are freed with cauterized scissors.

        If intact specimen extraction is to be performed, the lower midline trocar is then
exchanged for an entrapment sac device (15mm Endo Catch II, US Surgical, Norwalk, CT). The
specimen can then be placed into the deployed entrapment sac and delivered through a low
midline incision which incorporates the lower midline trocar. The extraction incision is then
closed and pneumoperitoneum is re-established to ensure that adequate hemostasis was obtained.
This is done at a pressure of 5mmHg to reveal bleeding that might be present at normal
insufflation pressures. Alternatively, the specimen can be morcellated. In these cases the
specimen must be placed in an impermeable entrapment sac (Lapsac® Cook, Bloomington, IN)
prior to morcellation. The 12mm trocar sites are then closed with the aid of a needle-point suture
passer after the pneumoperitoneum has been evacuated.

        Indications for laparoscopic radical nephrectomy include organ confined tumors (stage Tl
or T2) not amenable to nephron-sparing surgery. Some investigators have demonstrated that
even large masses (12cm) can be removed laparoscopically. Tumors spreading beyond Gerota's
fascia or involving the renal vein are technically challenging, but have been performed.
Laparoscopy has also provided a benefit to patients in the setting of metastatic disease.


Surgical Technique: Retroperitoneal Approach
         Positioning
         The patient is placed on the operating table in the standard flank position with the side of
pathology facing up. The top arm is draped over a padded Mayo stand or multiple pillows. An
axillary roll is placed. The bottom leg is flexed and bent while the top leg is straight. The kidney
rest is elevated and the table is flexed in order to maximize the working space between the ribs
and iliac crest (Picture 3).



                                                   20
Picture 3) The patient is placed in a full flank position with the kidney rest elevated and the table flexed.
This increases the distance between the costal margin and iliac crest, thus maximizing the working space.
        The skin between the iliac crest and the ribs should be taut on palpation. All pressure
points are padded and the patient is then secured to the table with tape at the shoulders, hips, and
legs. The table is rotated left and right to make sure the patient does not shift during the
procedure. The surgeon and camera assistant are positioned facing the patient's back. The scrub
nurse stands on the opposite side at the end of the table. (Fig 6)




                 Fig 6) Operating room setup for laparoscopic retroperitoneal renal surgery.
                                            From Pinto et al.25




                                                       21
Two monitors are used to provide all personnel with an unobstructed view. Alternatively,
the camera assistant can be replaced by a mechanical or robotic arm which can be fixed to the
operating table to hold the camera. The robotic arm is controlled by the surgeon through voice
commands, a hand control or a foot pedal.

        Access and Port Placement
        Three trocars are positioned in the anterior, mid, and posterior axillary lines.(Picture 4)




Picture 4) Trocar placement. The laparoscope is positioned in the mid-axillary line. A 12-mm port is placed
in the posterior axillary line. A 5-mm port is placed in the anterior axillary line.



        Initial entry into the retroperitoneal space is established off the tip of the 12th rib via the
open Hassan technique. A 1.5 cm transverse incision is made just anterior and inferior to the tip
of the 12th rib. The incision is carried down sharply through the posterior thoracolumbar fascia,
flank muscles and anterior thoracolumbar fascia.
        Upon entering the retroperitoneal space, the index finger is used to sweep the peritoneum
anteriorly and medially, developing a space between the psoas muscle anteriorly and Gerota’s
fascia posteriorly. (Fig 7)




                                                     22
Fig 7) The surgeon's finger is used to start the retroperitoneal dissection.
            A space behind the kidney is created to place the balloon dissector. See Pinto et al.25

The retroperitoneal space can then be created with balloon dissection (Fig 8A) and the balloon
tipped 10mm trocar (Fig 8B) provides a seal to establish the pneumoretropertioneum.

A)                                                                  B)




     Fig 8 A) The working space is created by balloon dissection. The kidney and peritoneum are
     displaced anteriorly exposing the renal hilum. B) Blunt-tipped trocar. The collar on the port
     slides down against the balloon tip to prevent loss of pneumoretroperitoneum. See Pinto et al.25

        Alternatively, retroperitoneal access can be obtained directly with a 12 mm laparoscopic
visualizing trocar. Once the characteristic fat of the retroperitoneum is seen, insufflation is
begun. The laparoscope is used to bluntly develop the working space.


Surgical Technique
        Renal dissection is carried out as was described previously, but without the need to
manipulate the peritoneal organs. The psoas muscle and ureter are the initial structures for
orientation, since the intraperitoneal structures are not seen. Maintaining orientation is important
throughout the procedure. This helps identify landmarks such as the psoas muscle, peritoneal


                                                      23
reflection, ureter, gonadal vein, and pulsations of the renal artery. Initial dissection is carried out
posteriorly to isolate the ureter and mobilize the lower pole.
        The peritoneal attachments anterior to the kidney are not divided in order to prevent the
kidney from falling into the working space. Blunt dissection along the ureter reveals the gonadal
vein. Theses structures are traced cephalad to the renal hilum. Dissection along the aorta or vena
cava will quickly bring the renal vessels into view. The renal artery is encountered first as it lies
posterior to the vein. Hilar ligation is performed as was described in the transperitoneal
approach. Once the vessels have been controlled, the anterior surface of the kidney is mobilized
from its peritoneal attachments. Care is taken not to enter the peritoneum and injure the adjacent
bowel and mesentery.

Post-Operative Management
         Prior to extubation the orogastric tube is removed. After routine observation in a post
anesthesia care unit, the patients are transferred to a non-monitored floor bed. In the morning of
the first post operative day, the urinary catheter is removed, routine blood work is performed,
ambulation is encouraged and oral liquids are started. The diet is then advanced as tolerated
throughout the day. Routine discharge is in the morning of post-operative day # 2 if blood work
and vital signs are stable, the patient is able to ambulate and tolerate oral diet.




                                                  24
III. Techniques and Indications:

       B) Laparoscopic Partial Nephrectomy
Surgical Technique: Transperitoneal / Retroperitoneal Approach
        Positioning and Port Placement
        Same as for laparoscopic nephrectomy. Additional instruments needed for laparoscopic
partial nephrectomy include: laparoscopic vascular clamps (Picture 5- bulldogs, Picture 6-
Statinksy), ultrasound with a laparoscopic transducer and topical hemostatic sealant.




      Picture 5) Laparoscopic bulldog clamps




    Picture 6) Laparoscopic Statinsky clamp

        Procedure
        When the renal mass is abutting the collecting system, and entry into a calyx is expected,
cystoscopy and placement of an open ended 5 French ureteral catheter is performed before
positioning. This aids in reconstructing the collecting system by retrograde injection of dilute
methylene blue. As in open surgery, dissection of the kidney is performed leaving a “cap”of fat
overlying the renal tumor. Intraoperative ultrasound aids in determining the tumor’s depth and
also to rule out any additional tumors.



                                                25
Exophytic masses that are attached by a narrow base and peripherally located can be
excised without the need for vascular occlusion. The capsule around the tumor is scored with
cautery, and the mass is removed with the aid of laparoscopic scissors or an ultrasonic scalpel.
Hemostasis is obtained with the aid of an argon beam coagulator and hemostatic sealants.
        Large, more broad-based or deeper tumors require interruption of renal blood flow. Prior
to clamping the renal hilum, a brisk diuresis is initiated with the use intravenous fluids and
osmotic diuretics. Mannitol (25 grams) is given when the surgeon begins to free up the renal
hilum allowing for increased perfusion of the renal parenchyma along with protection from free
radical deposition. The capsule around the mass is scored, and a laparoscopic bulldog clamp is
placed on the renal artery. Alternatively, a laparoscopic Statinsky vascular clamp can be used,
but this requires the placement of an additional trocar. Hilar clamping is performed to allow for a
bloodless field and therefore better visualization when resecting the tumor. The tumor is excised
with cold scissors to allow pathological assessment of the tumor’s margins. The tumor is excised
ensuring a rim of normal parenchyma around it. Any suspicious areas in the renal bed can be
biopsied and sent for frozen section analysis.
        Retrograde instillation of methylene blue via the ureteral catheter ensues that the
collecting system has not been entered. If it has, repair is performed with 2-O polyglactin suture.
Open vessels seen in the renal bed are suture ligated with 2-O polyglactin suture. A gelatin
matrix hemostatic sealant is applied and the kidney is reconstructed by reapproximating the renal
defect over surgicel bolsters (Picture 7) using O polyglactin sutures through the renal capsule.




                   Picture 7) Surgicel pledgets, approximately 1 cm wide and 4 cm
                   long, are prepared prior to tumor excision.

        The clamps on the renal vessels are removed and the kidney is observed to confirm it is
being well perfused and there is no active bleeding. Prior to exiting the abdomen, a closed
suction drain is left in the retroperitoneum behind the kidney.
        Cold ischemia is technically feasible laparoscopically, but not practical. Therefore when
operating under the time constraints of warm ischemia, it is important to be expeditious in your
approach to resection and reconstruction of the kidney.
        Indications for laparoscopic partial nephrectomy are as described for the open procedure.
Although initially indicated for tumors involving a solitary kidney or bilateral disease, nephron-
sparing surgery is now performed for tumors smaller than 4cm in the presence of a normal
contralateral kidney.




                                                  26
IV. Techniques and Indications:

       Robotic Assisted Nephrectomy and Partial Nephrectomy
        Nephron sparing surgery for complex renal tumors, such as multiple, endophytic, or hilar
tumors, may further preclude a laparoscopic approach for most urologists and may pose
challenges for even experienced laparoscopic surgeons. A renal mass with tumor thrombus may
require vascular suturing, which is also challenging for laparoscopic techniques. It is in these
cases that robotic renal surgery may be of assistance.
        Potential advantages include 3-dimensional stereoscopic vision, articulating instruments,
and scaled-down movements reducing tremor. The articulating instruments and increased
freedom of movement may also allow the surgeon to replicate well established open surgical
maneuvers more readily. This technique emulates both open and laparoscopic techniques of renal
cancer surgery. (Rogers et al. 2008)

        Surgical Technique: Transperitoneal Approach
        Positioning, Ports, and Docking:
        After intubation, orogastric tube placement and bladder catheterization, the patient is
positioned in modified lateral position. Full flank position may also be used. Following
abdominal insufflation with a Veress needle. Ports are placed as demonstrated in Picture 8. A
12mm periumbilical port is placed for the camera. Two robotic instrument ports are placed
approximately 8 cm from the camera in a wide “V” configuration centered on the renal tumor.
These ports may be shifted laterally and/or superiorly for patients with a large body habitus or
upper pole tumor location. A 12 mm assistant port is placed inferior to the camera port. An
optional 5mm assistant port may be placed above the camera port if needed. For docking (as
seen in Picture 9), the robot is brought in posteriorly at approximately a 20 degree angle toward
the head of the patient.




                      Picture 8) Port placement for left sided robotic renal surgery




                                                   27
Picture 9) daVinci robot is docked against the patient’s back at
                       approximately a 20 degree angle toward the head of the patient.



        Bowel Mobilization:
        A zero degree lens is used initially, but a 30 degree downward lens may also be utilized
as needed. Robotic instruments used include a Bipolar Maryland forceps, monopolar cautery
scissors, and needle drivers. The peritoneum is incised sharply along the Line of Toldt and the
bowel is mobilized medially using sharp and blunt dissection, developing the plane between the
anterior Gerota’s fascia and the posterior mesocolon. The bedside assistant maintains medial
counter-traction. Dissection is continued along the upper pole of the kidney to mobilize the
spleen or liver. We utilize robotic assistance from the beginning of bowel mobilization unlike
some reports of a “hybrid technique”, initially utilizing conventional laparoscopy to reflect the
bowel.

        Anatomical Landmarks and Hilar Dissection:
        Continued medial reflection of the bowel allows for exposure of the gonadal vessels and
the ureter. These structures are retracted anteriorly, exposing the underlying psoas muscle. Care
is taken not to strip the fascia from the psoas muscle. Dissection then proceeds towards the renal
hilum. The Maryland bipolar forceps are used to place the kidney on stretch and the renal hilar
vessels are dissected to allow access for clamp placement. Lateral renal attachments are left in
place to aid in countertraction. Venous branches can be ligated as needed for exposure

        Ultrasound and Tumor Exposure:
        A laparoscopic ultrasound probe is used to map the location and size of renal tumor(s)
and to confirm resection margins and depth during partial nephrectomy. Gerota’s fascia is
opened and the fat is cleaned off the renal capsule to expose the tumor(s). The use of color
Doppler may be used to identify adjacent vessels. The margin of resection is scored



                                                  28
circumferentially using monopolar cautery. In cases of nephrectomy and tumor thrombectomy, it
is used to determine the extent of tumor thrombus.

        Hilar Clamping, Tumor Excision and Renal Reconstruction during Partial
        Nephrectomy:
        For tumors that are endophytic or adjacent to the renal hilum, resection is done under
warm ischemia. The assistant clamps the renal hilar vessel(s) using laparoscopic bulldog
clamp(s) through the primary 12mm assistant port. Mannitol (12.5 gm) may be administered
intravenously prior to clamping. The tumor is resected along the previously scored margin using
cold resection with the robotic monopolar scissors. The Maryland bipolar forceps are used to
manipulate the tumor for exposure and to aid in dissection. The assistant uses suction to expose
and maintain visualization of the resection plane of the tumor. After excision, the tumor can be
placed beside the kidney or on top of the liver for later retrieval. If the status of the surgical
margin is in question, a biopsy and frozen section analysis may be performed.
        Hemostasis is achieved using a combination of cautery, hemostatic agents, and suturing.
A pre-placed ureteral catheter may be used to inject methylene blue to identify entry into the
collecting system. The robotic instruments are exchanged for needle drivers. A 3-0 Vicryl suture
on an RB-1 needle is used to achieve hemostasis and repair any previously identified entry into
the collecting system (a SH needle may also be used). Sutures may be secured with either
absorbable suture clips or by tying knots. Renal parenchymal defects are approximated over
surgicel bolsters using 2-0 Vicryl sutures on an SH needle (a 0-Vicry suture on a CT-1 needle
may also be used). A hemostatic agent is applied. Preplacing surgicel bolsters and sutures in the
abdomen may reduce warm ischemia time during earlier experience. The kidney is placed back
on stretch using the robotic needle driver and the hilar clamp is removed by the assistant.
Hemostasis is confirmed. A pre-placed lap pad may be used to apply pressure to the resection
site.
        The specimen is placed in a retrieval bag and removed through the primary assistant 12
mm port, enlarging the port site if needed. Gerota’s fascia is approximated over the defect using
a running 3-0 Vicryl suture on an SH needle. A drain is placed in the perinephric space. 26-54




                                               29
V.    Role of Targeted Systemic Therapy in Management of Advanced Clear
Cell and non-Clear Cell Kidney Cancer (Jeffrey A. Sosman, M.D.)


Introduction
Therapy of advanced Renal Cell Carcinoma (RCC) has gone through a major evolution over the
past few years. Previously, patients with RCC were considered for high dose Interleukin-2
therapy, if young and healthy and IFN-alpha for the less robust or elderly patient. A relatively
small subset of patients did benefit from cytokine therapy, but far too few to effect median
progression-free (PFS) or overall survival (OS)55,56. The great majority of patients only suffered
the toxicity of treatment. This all began to change following the remarkable work of Drs. Kaelin,
Maxwell, Linehan and others who helped define the role of the VHL gene and the consequence
of loss of its function.57 The generation of an antibody to the vascular endothelial growth factor,
VEGF (anti-VEGF; Bevacizumab) molecule provided an initial tool to interrupt the downstream
effects of VHL (Von Hippel Lindau) loss. The stabilization of the HIF-1a and HIF-2a molecules
with the subsequent over expression of VEGF, PDGF, EGFR, CAIX, EPO, Glut1 etc was key to
the malignant phenotype of clear cell RCC. This is a critical pathway not simply in genetic
syndromes with Von Hippel Lindau, but in at least the majority of patients with sporadic clear
cell RCC.

In the initial report by Yang et al., anti-VEGF was shown to increase PFS >2 fold in a cytokine
(IL-2) refractory population even though its objective response rate was only 10%.58 However,
it was clear that even those without objective clinical responses had a far different clinical course
than those on placebo with many having prolonged stable disease or small degrees of regression
not meeting the 30% RECIST threshold of an objective response. A reassessment of meaningful
clinical responses needed to be performed the first time there may have been convincing data
that stable disease was an indication of benefit in RCC. In the interim, a number of multi-
targeted tyrosine kinase inhibitors (TKI) that targeted the VEGFR2 tyrosine kinase as well as a
variety of other receptor signaling pathways, have been evaluated and some approved for therapy
of advanced RCC. These orally administered drugs were also able to inhibit other receptors
including VEGFR1, VEGFR3, PDGF, c-kit, and RAF to name a few. The initial two agents
receiving the most attention and most rapidly approved by the FDA were BAY43-9006
(Sorafenib) Nexavar and SU011248 (Sunitinib) Sutent.59 While their spectrum of kinase targets
was overlapping, there were some key differences. Sorafenib inhibited RAF, while Sunitinib was
a more potent and effective inhibitor of VEGFR2. (See Table 1)




                                                 30
Table 1: Phase II/III Trials of Targeted Agents in Untreated RCC patients
                       Phase
                             Pt                                                        Response       Median    Median
Drug                   of           Risk factors                    Nephrectomy
                             Number                                                    Rate           PFS       OS
                       Study

                               375         Favorable(34%)                              39%            11 mos    NA
Sunitinib vs IFN       III                                   90%
                               375         Intermediate(56%)                           8%             5.1 mos   NA
Bevacizumab +                  327         Favorable (28%)                             31%            10.2mos   NA
                       III                                   100%
IFN vs IFN                     322         Intermediate(56%)                           12%            5.4 mos   NA
Temsirolimus                   209                                                     8.6%           5.5mos    10.9mo
                                           Poor Prognosis
Temsirolimus+IFN III           210                                  67%                8.1%           4.7mos    8.4 mo
                                           (74% MSKCC)
Vs IFN                         207                                                     4.8%           3.1mos    7.3 mo
Sorafenib                      97          Favorable                                   NA(68%)*       5.7mos    NA
                       II                                           95%/98%
Vs IFN                         92          Intermediate                                NA(39%)*       5.6mos    NA
       Number in parenthesis represents % of patients with some degree of tumor shrinkage, not an objective
       response rate



       A) Sorafenib
               Initially, Sorafenib demonstrated prolonged progression-free survival in cytokine
       refractory patients in a phase II randomized trial where stable disease patients at 12 weeks were
       randomized to continue Sorafenib or switched to placebo (randomized discontinuation).59 Those
       patients having switched to placebo had a significantly less PFS than those on Sorafenib, when
       the placebo patients who progressed were rechallenged with Sorafenib, stable disease could be
       re-established. This was confirmed and proven in a large phase III trial of Sorafenib versus
       placebo in cytokine-refractory patients where again as in the phase II trial there was a doubling
       in progression-free survival from 2.8 to 5.5 months9. Unfortunately this study was then
       unblinded (per recommendations of the FDA) and overall survival results were confounded by
       the significant cross-over of patients from placebo to Sorafenib. It is worth pointing out that in
       both of these studies the objective response rate of Sorafenib was < 10% while many patients had
       smaller degrees of regression and prolonged stable disease.

               Later after its approval a relatively small, 189 patient, randomized phase II trial of
       Sorafenib or IFN in therapy-naïve patients was conducted. Surprisingly, there was no advantage
       to the Sorafenib arm.60 This was unexpected based on the second line results with Sorafenib in
       cytokine-refractory patients. While a low objective response rate could have been expected, the
       lack of any improvement in patients’ PFS with a median of 5.7 months, was a surprise. The trial
       also examined the effect of cross over to Sorafenib and dose escalation of Sorafenib in patients
       who progressed on their initial therapy. Patients progressing on IFN, could receive Sorafenib at
       standard doses of 400mg BID. This group of patients did exhibit some stabilization of their
       disease for 5.7 months and some degree of tumor shrinkage in 75% of patients. Those who
       progressed on Sorafenib could be dose escalated to 600mg BID and among these 44 patients, a
       4.1 months median PFS and tumor shrinkage in 44% were observed. For all practical purposes,
       this large randomized phase II trial signaled the end of Sorafenib as a front line therapy in the


                                                           31
majority of patients. Larger, single agent, upfront trials with Sorafenib at standard doses, are
unlikely to be explored further.

B) Sunitinib
        The pathway to Sunitinib approval was quite different. Initial phase II studies did
demonstrate a high response rate of 35-45% in cytokine-refractory patients, as well as prolonged
progression-free survival. Following this result, Sunitinib was directly compared to IFN-alpha in
therapy naïve patients. This large 750 patient trial demonstrated a remarkable objective response
rate for Sunitinib that remained very high at 40%, while IFN's response rate was <10%8.
Furthermore, the median progression-free survival was 11 months, more than double that of
IFNα. The difference in overall survival did not meet significance due to the lack of events at the
time of the analysis. Again the survival analysis is confounded by patient crossover from IFN to
Sunitinib as allowed in the protocol once Sunitinib was approved by the FDA.


C) Bevacizumab
         Following the approval of both MTKI, the results of several phase III trials featuring
Bevacizumab (anti-VEGF) have been reported. In a large study now published in Lancet by
Escudier, et al., IFN alone was compared to IFN with anti-VEGF in a population of therapy-
naïve RCC patients.61 The results showed a striking improvement in response rate (31% vs 7%)
and progression-free survival (10.2 mos vs 5.4 mos). The response rate was better than seen in
both the initial Yang trial in cytokine refractory patients (10%) and the 13% in the randomized
phase II trial of anti-VEGF +/- erlotinib (EGFR TKI). It is possible that the addition of IFNα to
anti-VEGF led to a better response rate and improved the progression-free survival of anti-
VEGF. But there are no more definitive results to either support or counter this argument.
Finally, limited release results from a trial conducted by CALGB show there were further
supporting data to the benefit of Bevacizumab plus IFN. The trial represented another phase III
trial of IFN +/- anti-VEGF. The combined arm (anti-VEGF + IFN) demonstrated an improved
response rate and progression-free survival compared to IFN alone. Final results are still
pending.

Issues with phase III trials in front line therapy
         When reviewing the important trials with Sorafenib, Sunitinib, and Bevacizumab, there
are several important issues needing to be addressed. First, patients on all of these studies were
either intermediate or low-risk based on the MSKCC criteria62. They had 0-2 of (anemia,
hypercalcemia, high LDH, lower performance status (<70% Karnofsky criteria) or either
within 1 year of diagnosis of RCC or without nephrectomy) the five criteria. Three or more
of these criteria were indicative of poor prognosis. In previously evaluated patients at MSKCC,
this criteria could separate patients with a 20 month median (low risk, favorable prognosis)
overall survival to as low as a 4 month median survival for those high risk (poor prognosis)
patients. The entry criteria for all of the above studies also required clear cell histology. All
patients on the Bevacizumab plus IFN trial had nephrectomy (as per entry criteria), while over
90% had prior resection of their primary on the Sunitinib trial. There were a small number of
poor prognosis patients entered on both trials, 6% on the Sunitinib trial and 8% on the
Bevacizumab + IFN trial. Both the front and second line trials with Sorafenib enrolled patients
with intermediate to low risk and nearly 90% had nephrectomy. The results in the poor


                                                 32
prognostic patients were dismal but PFS may have been improved from 1.2 months to 3.7
months in those receiving Sunitinib. Not even this small effect was seen in the Bevacizumab+
IFN trial where poor prognosis patients had a 2.1-2.2 median progression free survival regardless
which arm they were enrolled on.

D) Temsirolimus
         In phase I and II clinical trials, RCC patients appeared to derive a benefit of CCI-779,
Temsirolimus, even though the response rates were less than 10%. Patients improved with less
bone pain, improved appetite and weight gain. Temsirolimus is a mTOR (mammalian target of
rapamycin) inhibitor. mTOR is downstream from PI-3/Kinase/ Akt. It represents a pathway that
can modulate protein synthesis (translation) including that of HIF-1a and HIF-2a. The phase II
trial demonstrated that patients with poor prognostic factors had the greatest benefit. Benefit was
based on a major extension in progression-free and overall survival in the poor prognosis
group.63 Overall survival was much better than expected for this poor prognosis patient group.
This led to a phase III trial comparing Temsirolimus versus IFN in poor prognostic patients (3 or
more of 5 criteria in MSKCC). The trial did get slightly modified to increase poor accrual by
adding one more poor prognostic factor, multiple disease sites (by organ involvement). Patients
needed 3 or more of the 6 risk factors for entry. There were no restrictions on histology and
requirements for nephrectomy. About 20% had non-clear cell histology and about 33% had their
primary in place and were enrolled within their first year of diagnosis. The trial was an ambitious
effort in a poor prognostic group of patients who in the past would likely not have been treated
with cytokines due to their overall performance status. The trial included 3 arms; one with IFN
alone; one with Temsirolimus; and the other a combination of both IFNa plus Temsirolimus 64.
The doses of Temsirolimus were attenuated to allow concomitant IFNa therapy. The trial’s
primary endpoint was overall survival, with progression-free survival only a secondary endpoint.
Overall survival was increased from a median of 7.3 to 10.9 months with an overall hazard ratio
of .73 and the p value=0.0069. PFS was improved in both the Temsirolimus arm and the IFN
plus Temsirolimus arm, but those in the combined arm did not get any improvement in overall
survival. This may have to do to the extensive dose modification of when IFN and Temsirolimus
were given together. This result clearly established Temsirolimus as the standard therapy for
poor prognostic patients. Overall response rates in all three arms were under 10%, further
evidence that objective responses were a poor indicator of treatment benefit from drugs in RCC.
Several additional analyses were done retrospectively on pre-treatment patient characteristics and
clinical outcome. For those 74% of the overall enrollment who did qualify under the MSKCC
model as a poor prognostic group (3 or more of 5 MSKCC criteria) the benefit from
Temsirolimus was most prominent. Most of the benefit was apparently concentrated within this
group of patients. Furthermore, about 20% of patients had histology of their tumor different from
the typical clear cell. Papillary renal cancer makes up the bulk of these non-clear cell cancers.
Others include chromophobe and sarcomatoid (high grade IV) that make up a small number of
cases. In those patients with non-clear cell histology the benefit was especially prominent as is
described below in non-clear cell RCC. This was unique to this trial of Temsirolimus since the
other upfront trials excluded these patients and were restricted to clear cell RCC alone.




                                                33
Toxicities from the targeted agents (See Table 2)
        In addition to overall survival, progression-free survival, and objective response rate,
toxicity, and patient tolerance is of critical importance when considering treatment with this class
of agents. This is especially true, since the nature of these agents is to continue their
administration indefinitely or until progression. Complete responses are very rare and much of
therapy is administered with the intent to maintain disease control. Both Sunitinib and Sorafenib
have the most overlapping spectrum of toxicities as might be expected based on the targets of
their action.
                            Table 2: Toxicities from Targeted Agents
Toxicities       HTN Proteinuria Vascular                 Stomatitis HFS Rash Diarrhea Fatigue
                                        Complications
Sunitinib        X                                        X            X      x      X         X
Sorafenib        X                                        x            X      X      X         X
Temsirolimus                                              x                   X                X
Bevacizumab X            X              X
X Common and significant problem
x Less common and/or infrequent problem


Sunitinib toxicity
       Sunitinib can cause fatigue, hypertension, a functional (symptomatic with minimal
findings on examination) stomatitis, diarrhea, and some hand-foot syndrome. Most common
grade 3 or 4 toxicities have been due to fatigue and hypertension, though hypertension in general
can be well controlled with medications. At times multiple anti-hypertensive agents are required
for adequate blood pressure control. Hypothyroidism is becoming more appreciated as a
frequently associated side effect. Thyroid functions should be followed throughout treatment.
Sometimes the fatigue from Sunitinib is due to thyroid dysfunction and this can be easily
addressed with hormone replacement. A less frequent but increasingly recognized toxicity is a
decline in cardiac ejection-fraction. While declines below normal range or greater than 10% of
baseline are relatively common at 10-15%, symptoms of congestive heart failure is seen in less
than 5% of patients. Obtaining baseline ejection fraction and subsequent follow-up cardiac
evaluations are now recommended even in the absence of symptoms.

Sorafenib toxicity
         Sorafenib toxicities include many of those seen with Sunitinib but there is a slight but
recognizable change in the spectrum of them. Hypertension is common as with Sunitinib, but
fatigue is infrequently a dose limiting toxicity. Diarrhea is experienced in up to 40% of patients
but is grade 3 or 4 in only several percent. Most bothersome has been the hand foot syndrome
that is observed in up to 30% of patients. While severe desquamation of the hands and feet is
uncommon, symptomatic callus formation with neuropathic pain is frequently observed. Patients
can have trouble with walking and use of their hands. This toxicity is readily reversible upon
suspending drug administration, but if the symptoms worsen while drug continues to be given
then recovery can be quite slow. Sorafenib can also induce a number of different rashes from a
diffuse macular, papular, pruritic rash to a localized acneform rash similar to that associated with
EGFR tyrosine kinase inhibitors or antibodies. It can also cause multiple furuncles and



                                                34
carbuncles in and around the axillas or the inguinal regions. These can become secondarily
infected and required drainage and antibiotics.

Bevacizumab toxicity
        Bevacizumab is very well tolerated on a daily basis. Minor fatigue and myalgias can be
associated with intravenous administration for 24-48 hours afterwards. Patients frequently
experience hypertension and proteinuria. Blood pressure control is important since lack of
control can lead to hypertensive crisis. Proteinuria must be monitored closely and in general is
very reversible. Recommendations have become more lax in terms of the threshold for
proteinuria in deciding to hold Bevacizumab. At the present time, 3.5 gm of protein in a 24 hour
urine is the limit for continuing Bevacizumab dosing. Otherwise the dose is held until protein in
urine has dropped below the threshold. Bevacizumab is generally safe and well tolerated. It must
be recognized that Bevacizumab can significantly slow wound healing and even lead to wound
dehiscence. This must be considered around procedure and any major surgery. In general
Bevacizumab should be held two weeks prior to surgery and at least 4 weeks following surgery
to allow healing to take place optimally. However there are much more infrequent toxicities of a
very serious nature. These include vascular events, especially arterial thrombotic and venous
thromboembolic , hemorrhage, multifocal leukoencephalopathy associated with poorly
controlled hypertension, gastrointestinal perforation, and fistula formation.

Temsirolimus toxicity
        Finally, Temsirolimus is tolerated quite well. Some degree of asthenia (fatigue) is
relatively common. However, this can be difficult to attribute to either disease or therapy.
Additionally, patients have some degree of pancytopenia with anemia being most prominent.
Secondary infection or bleeding is very rare. Some mouth ulcerations and rashes can also be
seen. An untoward toxicity relatively unique to this agent is hyperglycemia, hypertriglyceremia,
and hyperlipidemia. Pancreatitis may occur secondary to the elevated tryglycerides. While
therapy can be cumbersome with its weekly intravenous schedule, its overall toxicity is mild
even in the overall more debilitated patients who have been studied.

Alternate Doses and Schedules
         For two of the agents, alternative dosing and scheduling have been attempted. Sunitinib
can be given continuously at 37.5mg po daily. This schedule avoids the break in treatment for 2
weeks where conditions may favor regrowth. However, the lower dose, continuous schedule was
not superior and the overall response rate appeared lower than the standard 50mg daily for 4 of 6
weeks. This schedule was not directly compared to the standard schedule in a randomized trial.
Additionally, toxicity was similar with the continuous dosing schedule. Attempts have been
made at dose escalation of Sorafenib. There was some evidence from the randomized phase II
trial of Sorafenib that those who progressed on 400mg BID could be dose escalated to 600 mg
BID. Amato and his colleague have attempted to follow a very aggressive dose escalation in
treatment naïve patients. In 44 patients, 41 were able to be escalated to 600mg BID after 1 month
and then 32 of these patients were able to be increased to 800mg BID after 2 months65. There
was a very high response rate of 55% with 16% complete responses. Though the median PFS
was just 8+ months and overall survival was at 11.5 months. This is surprisingly short
considering the high response rate and suggests that these are short clinical responses that may
be limited by toxicities. This must be reproduced in a multi-center phase II study, but if positive



                                                35
it would be quite significant and ways to ameliorate Sorafenib toxicities would take on even
more importance. Until then these results should be looked at with great caution and only
considered in a trial setting.


Therapy in Patients Who Have Failed Front Line Anti-angiogenesis Therapy
        There is little information to guide therapy in patients failing their initial treatment. One
study has provided some direction in patients who have failed Bevacizumab66. These patients
with progressive disease while on Bevacizumab were then treated with Sunitinib. Therapy had to
be initiated within several months of stopping Bevaciumab treatment. Of 61 patients there was
an objective response rate of 23% with a median progression-free survival of 30.4 weeks (7.4
mos) and overall survival of 47.1 weeks (10.9 months). Of the 57 patients with post-treatment
tumor evaluations 51 (84%) demonstrated some degree of tumor regression. Patients who had
failed to have any prior response to Bevacizumab were among those who demonstrated objective
responses to Sunitinib (11/14 patients with PR). To date, this represents the most mature study of
second line therapy.

        Rini, et al. reported on a cohort of 62 patients who had failed Sorafenib and were
subsequently treated with another multi-targeted kinase inhibitor, Axitinib with potent activity
against all three VEGFR1, 2,and 3.67 Many of these patients had not only failed Sorafenib, but
also Sunitinib, cytokines, and chemotherapy. Of 62 patients, 21% experienced a partial clinical
response and 34% had stable disease for at least 3 months. Hypertension as was previously
observed in frontline studies was the most problematic side effect from Axitinib.

        Finally, Escudier has reported on 90 patients from France who had previously received
Sorafenib and following progression were switched to Sunitinib (68 patients) and 22 patients
who had failed Sunitinib and were then switched to Sorafenib therapy. Among those 68 patients
who were switched to Sunitinib there was a 15% objective response rate with an additional 51%
with stable disease. The median PFS was 25 weeks. For the 22 patients who had failed Sunitinib
and switched to Sorafenib, there was a 9% response rate with stable disease in 55% and median
PFS of 17 weeks. While this was only a retrospective study it does support the lack of complete
cross resistance for these two drugs even with their similar target profile.

Combination Targeted Therapy in Advanced RCC
        A number of studies have been reported combining several of the active targeted agents
including Sorafenib or Sunitinib with Bevacizumab and Temsirolimus with either Bevacizumab,
Sorafenib or Sunitinib. While most of these studies enrolled small numbers of patients and
included several dose levels, there have been some common findings. In general toxicity of the
two agents appears to be exacerbated. There has been an increase in known toxicities such as
hand-foot syndrome, hypertension, or proteinuria. In some cases, new toxicities have occurred
such as a microangiopathy associated with hemolysis, thrombocytopenia, renal insufficiency and
mental status changes. That has been recently observed with Sunitinib and Bevacizumab
combination. While some combinations appear to allow full doses of both agents (Bevacizumab
and Temsirolimus) most of the combinations required dose reduction of either one or both of the
agents. Combinations of Sorafenib and Bevacizumab have required significant dose reductions
of both agents. Activity in many cases appears to be preserved but still no formal phase II studies



                                                 36
have been undertaken with any of the combinations. A recently opened randomized phase II trial
will explore three of the combinations of Sorafenib with Bevaciumab, Sorafenib with
Temsirolimus, Bevacizumab with Temsirolimus, and as a control, Bevacizumab alone.
Additionally, another phase III trial is being planned to compare Bevacizumab with
Temsirolimus or with IFN. In general, there remains many questions about combination therapy,
especially in the face of toxicity and requirements for lower doses of the agents when they are
combined. So far complete responses have been as rare as with single agents. It may be in the
future that sequential therapy will be considered the best approach to treatment as it is frequently
done today. Only through well performed trials will we gain this information.

Use of Targeted agents in the treatment of Papillary Renal Cell Carcinoma (See Table 3)
         The role of HIF and its hypoxia induced gene expression program in papillary RCC is not
understood. In the past most medical urologists, believed these malignancies responded to
chemotherapy based regimens. This was never adequately studied to draw any conclusions.
Probably the most convincing information about treatment efficacy in papillary RCC comes from
the front line Temsirolimus trial as described above. In this trial out of 626 patients about 19%
had non-clear cell histology. Of those where histology was better described 75% had papillary
RCC as would be expected. In the two arms of Temsirolimus versus IFN there were 73 patients
with non-clear cell histology. In a striking result, the progression-free survival was 7.0 versus
1.4 months favoring the Temsirolimus arm and overall survival was more than doubled in those
receiving Temsirolimus with OS of 11.6 months versus 4.3 months for IFN–treated patients. This
data has a number of problems; the number of patients is small and the actual with papillary
RCC is not definite, but assuming the arms are balanced and most are papillary cancers, these
results suggest Temsirolimus is an active drug in papillary RCC.
         Finally, in a retrospective report recently published from 5 centers in the US and France,
the clinical outcomes of Sorafenib and Sunitinib in patients with papillary RCC was reported.
Forty-one of the 53 patients had papillary RCC and the response rate was only 4.8% (2/41) and
both received Sunitinib. The median PFS for papillary RCC treated with Sunitinib was 11.9
months (13 patients) while for the other 28 patients treated with Sorafenib the median PFS was
5.1 months. Few patients had central pathology review. While the data on Sunitinib is limited
with only 13 patients, there appears to be some activity (2/13 patients with PR) and PFS was
11.9 months. On the other hand, therapy with Sorafenib had only a median PFS of 5.1 months
and no clinical responses in the 28 patients. It is obvious further information is needed to make
any definite decisions on therapy with either Sunitinib, Sorafenib or even Temsirolimus. It is
critical to perform large enough multicenter studies with only papillary RCC to better gauge
effects of any of the above agents.




                                                 37
Table 3: Experience with Targeted Agents in Papillary Renal Cell Carcinoma
 Drug            Histology          Patient   Response        Median PFS Median OS
                                    Number    Rate
 Sorafenib         Papillary              28             0/28            5.1 mos          NA

 Sunitinib         Papillary              13             2/13            11.9 mos         NA
 Temsirolimus Non-clear cell#             37             NA              7.0 mos          11.6 mos
 Vs IFN       75% Papillary               36             NA              1.4 mos          4.3 mos
# Patients all had poor prognostic factors based on MSKCC and Multiple Organ Sites (>3/6 risk factors)




Summary (See Table 4)
         Great progress has been made in a few years as shown in Table 4. Frontline data supports
the use of either Sunitinib alone or Bevacizumab with IFN in patients with favorable or
intermediate prognostic factors. The role of nephrectomy is not known, but almost all patients
enrolled onto previous studies did have nephrectomies. Those patients with poor prognostic
factors should probably receive Temsirolimus. Data on Sunitinib is simply not very convincing
at this point but there very well could be activity comparable to Temsirolimus. Second line
cytokine failures would be appropriate candidates for Sorafenib based on its prolongation in
median PFS. Finally those who have failed front line targeted therapy have very little guidance
about what treatment should be given next. Bevacizumab failures can receive a significant
benefit from Sunitinib and patients who fail one of the MTKI, Sorafenib or Sunitinib can respond
with objective clinical responses or stable disease from the other agent. However, this is an area
which still requires extensive study. Lastly, patients who are young, healthy, have good organ
function, an excellent performance status, and access to an experienced IL-2 center may be
considered for high dose Interleukin-255 . It is the only treatment which offers a chance for cure
and very prolonged disease free periods. While this number is less than 10%, because of its
impact on these patients is so great, some may consider it for frontline therapy and targeted
agents after failing IL-2. We have impacted on many more patients that any of the cytokines ever
did. While few studies show an overall benefit except for Temsirolimus, those of us taking care
of patients have experienced living for a number of years with disease control where in the past
their survival was measure in months to a year. This impact will become more clear with time.
Hopefully over that time we will gain further insight how to use these agents.




                                                    38
Table 4: Potential Standards for Advanced Stage Clear Cell RCC
               Setting                                 Therapy
First-Line    Good or intermediate     Sunitinib           High-dose IL-2
 Therapy             risk*                    Bevacizumab? With IFN?

                   Poor risk*                 Temsirolimus/ Sunitinib

 Second-         Prior cytokine                       Sorafenib
  Line           Prior VEGFR                        Temsirolimus??
 Therapy           inhibitor
             Prior mTOR inhibitor                  No data available




                                         39
References

 1. Jemal, A., Siegel, R., Ward, E., Murray, T., Xu, J., and Thun, M. J.: Cancer statistics, 2007.
           CA Cancer J Clin, 57: 43, 2007.

 2. Linehan, W. M., Bates, S. E., and Yang, J. C.: Cancer of the Kidney. In: Cancer: Principles
          and Practice of Oncology 7th ed., 7th Edition ed. Edited by DeVita, S. E., Hellman,
          S., and Rosenberg, S. A.: Philadelphia: Lippincott Williams & Wilkins, chapt. 30,
          pp 1139-1168, 2005.

 3. Linehan, W. M., Vasselli, J., Srinivasan, R., Walther, M. M., Merino, M. J., Choyke, P. et
          al.: Genetic Basis of Cancer of the Kidney: Disease-Specific Approaches to
          Therapy. Clin Cancer Res, 10: 6282S, 2004.

 4. Linehan, W. M. and Zbar, B.: Focus on kidney cancer. Cancer Cell, 6: 223, 2004.

 5. Linehan, W. M., Walther, M. M., and Zbar, B.: The genetic basis of cancer of the kidney. J
          Urol, 170: 2163, 2003.

 6. Chen, F., Kishida, T., Yao, M., Hustad, T., Glavac, D., Dean, M. et al.: Germline mutations
           in the von Hippel-Lindau disease tumor suppressor gene: correlations with
           phenotype. Hum Mutat, 5: 66, 1995.

 7. Gnarra, J. R., Tory, K., Weng, Y., Schmidt, L., Wei, M. H., Li, H. et al.: Mutations of the
           VHL tumour suppressor gene in renal carcinoma. Nat Genet, 7: 85, 1994.

 8. Motzer, R. J., Hutson, T. E., Tomczak, P., Michaelson, M. D., Bukowski, R. M., Rixe, O.
          et al.: Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J
          Med, 356: 115, 2007.

 9. Escudier, B., Eisen, T., Stadler, W. M., Szczylik, C., Oudard, S., Siebels, M. et al.:
           Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med, 356: 125,
           2007.

10. Motzer, R. J., Hudes, G. R., Curti, B. D., McDermott, D. F., Escudier, B. J., Negrier, S. et
          al.: Phase I/II trial of temsirolimus combined with interferon alfa for advanced renal
          cell carcinoma. J Clin Oncol, 25: 3958, 2007.

11. Haas, N. B. and Atkins, M. B.: A New Adjuvant Trial is Born: E2805 to Study Effects of
           Sorafenib, Sunitinib after Nephrectomy. Kidney Cancer Journal, 4: 9, 2007.

12. Zbar, B., Tory, K., Merino, M. J., Schmidt, L., Glenn, G. M., Choyke, P. et al.: Hereditary
           papillary renal cell carcinoma. J Urol, 151: 561, 1994.




                                               40
13. Lubensky, I. A., Schmidt, L., Zhuang, Z., Weirich, G., Pack, S., Zambrano, N. et al.:
          Hereditary and sporadic papillary renal carcinomas with c-met mutations share a
          distinct morphological phenotype. Am J Pathol, 155: 517, 1999.

14. Schmidt, L., Duh, F. M., Chen, F., Kishida, T., Glenn, G. M., Choyke, P. et al.: Germline
          and somatic mutations in the tyrosine kinase domain of the MET proto-oncogene in
          papillary renal carcinomas. Nat Gen, 16: 68, 1997.

15. Schmidt, L., Junker, K., Weirich, G., Glenn, G., Choyke, P., Lubensky, I. et al.: Two North
          American families with hereditary papillary renal carcinoma and identical novel
          mutations in the MET proto-oncogene. Cancer Res, 58: 1719, 1998.

16. Pavlovich, C. P., Walther, M. M., Eyler, R. A., Hewitt, S. M., Zbar, B., Linehan, W. M. et
           al.: Renal tumors in the Birt-Hogg-Dube syndrome. Am J Surg Pathol, 26: 1542,
           2002.

17. Nickerson, M. L., Warren, M. B., Toro, J. R., Matrosova, V., Glenn, G. M., Turner, M. L.
           et al.: Mutations in a novel gene lead to kidney tumors, lung wall defects, and
           benign tumors of the hair follicle in patients with the Birt-Hogg-Dube syndrome.
           Cancer Cell, 2: 157, 2002.

18. Schmidt, L. S., Nickerson, M. L., Warren, M. B., Glenn, G. M., Toro, J. R., Merino, M. J.
          et al.: Germline BHD-mutation spectrum and phenotype analysis of a large cohort
          of families with Birt-Hogg-Dube syndrome. Am J Hum Genet, 76: 1023, 2005.

19. Grubb, R. L., III, Franks, M. E., Toro, J., Middelton, L., Choyke, L., Fowler, S. et al.:
           Hereditary leiomyomatosis and renal cell cancer: a syndrome associated with an
           aggressive form of inherited renal cancer. J Urol, 177: 2074, 2007.

20. Wei, M. H., Toure, O., Glenn, G. M., Pithukpakorn, M., Neckers, L., Stolle, C. et al.:
          Novel mutations in FH and expansion of the spectrum of phenotypes expressed in
          families with hereditary leiomyomatosis and renal cell cancer. Journal of Medical
          Genetics, 43: 18, 2006.

21. Merino, M. J., Torres-Cabala, C., Pinto, P., and Linehan, W. M.: The morphologic
          spectrum of kidney tumors in hereditary leiomyomatosis and renal cell carcinoma
          (HLRCC) syndrome. Am J Surg Pathol, 31: 1578, 2007.

22. Zbar, B., Glenn, G., Merino, M., Middelton, L., Peterson, J., Toro, J. et al.: Familial renal
           carcinoma: clinical evaluation, clinical subtypes and risk of renal carcinoma
           development. J Urol, 177: 461, 2007.

23. Pinto, P. A., Chien, G. W., and Shalhav, A. L.: Laparoscopic renal surgery., 2nd ed
            Hamilton, Ontario: B.C. Decker, Inc., 2007.

24. Williams, C. R. and Pinto, P. A.: Laparoscopic partial nephrectomy. In: Atlas of
           laparoscopic retroperitoneal surgery, In Press ed. Edited by Bishoff, J. T. and
           Kavoussi, L. R.: Milan: Springer, chapt. 2nd ed., 2017.


                                                41
25. Pinto, P. A. and Jarrett, T. W.: Renal Surgery in Adults. In: Retroperitoneoscopy and
            Extraperitoneal Laparoscopy in Pediatric and Adult Urology. Edited by Caione, P.,
            Kavoussi, L. R., and Micali, F.: Milan: Springer, 2003.

26. Clayman, R. V., Kavoussi, L. R., Soper, N. J., Dierks, S. M., Meretyk, S., Darcy, M. D. et
          al.: Laparoscopic nephrectomy: initial case report. J Urol, 146: 278, 1991.

27. Gill, I. S., Schweizer, D., Hobart, M. G., Sung, G. T., Klein, E. A., and Novick, A. C.:
             Retroperitoneal laparoscopic radical nephrectomy: the Cleveland clinic experience.
             J Urol, 163: 1665, 2000.

28. Micali, S., Caione, P., Virgili, G., Capozza, N., Scarfini, M., and Micali, F.:
            Retroperitoneal laparoscopic access in children using a direct vision technique. J
            Urol, 165: 1229, 2001.

29. Licht, M. R. and Novick, A. C.: Nephron sparing surgery for renal cell carcinoma. J Urol,
           149: 1, 1993.

30. Provet, J., Tessler, A., Brown, J., Golimbu, M., Bosniak, M., and Morales, P.: Partial
            nephrectomy for renal cell carcinoma: indications, results and implications. J Urol,
            145: 472, 1991.

31. Hafez, K. S., Fergany, A. F., and Novick, A. C.: Nephron sparing surgery for localized
           renal cell carcinoma: impact of tumor size on patient survival, tumor recurrence and
           TNM staging. J Urol, 162: 1930, 1999.

32. Ono, Y., Kinukawa, T., Hattori, R., Yamada, S., Nishiyama, N., Mizutani, K. et al.:
          Laparoscopic radical nephrectomy for renal cell carcinoma: a five-year experience.
          Urol, 53: 280, 1999.

33. Thomas, M. A., Rha, K. H., Ong, A. M., Pinto, P. A., Montgomery, R. A., Kavoussi, L. R.
         et al.: Optical access trocar injuries in urological laparoscopic surgery. J Urol, 170:
         61, 2003.

34. Finelli, A. and Gill, I. S.: Laparoscopic partial nephrectomy: contemporary technique and
             results. Urologic Oncology, 22: 139, 2004.

35. Thompson, T., Ng, C. F., and Tolley, D.: Renal parenchymal hemostatic aids: glues and
         things. Current Opinions in Urology, 13: 209, 2003.

36. Tsivian, A. and Sidi, A. A.: A simple and reliable hemostatic technique during partial
           nephrectomy. Urol, 63: 976, 2004.

37. Allaf, M. E., Bhayani, S. B., Rogers, C., Varkarakis, I., Link, R. E., Inagaki, T. et al.:
           Laparoscopic partial nephrectomy: evaluation of long-term oncological outcome. J
           Urol, 172: 871, 2004.




                                               42
Cancer renal
Cancer renal
Cancer renal
Cancer renal

Weitere ähnliche Inhalte

Was ist angesagt?

Medical Students Who Want to Become PCPs Are Reconsidering Their Options
Medical Students Who Want to Become PCPs Are Reconsidering Their OptionsMedical Students Who Want to Become PCPs Are Reconsidering Their Options
Medical Students Who Want to Become PCPs Are Reconsidering Their OptionsMedical Transcription Service Company
 
Best Practices: Needs Assessment Process, Data/Metric Tracking, and Survivors...
Best Practices: Needs Assessment Process, Data/Metric Tracking, and Survivors...Best Practices: Needs Assessment Process, Data/Metric Tracking, and Survivors...
Best Practices: Needs Assessment Process, Data/Metric Tracking, and Survivors...Academy of Oncology Nurse Navigators, Inc.
 
Medicine grand rounds
Medicine grand roundsMedicine grand rounds
Medicine grand roundsusacme
 
Pancreatic-Effective-Practices-in-Pancreatic-Cancer-Programs
Pancreatic-Effective-Practices-in-Pancreatic-Cancer-ProgramsPancreatic-Effective-Practices-in-Pancreatic-Cancer-Programs
Pancreatic-Effective-Practices-in-Pancreatic-Cancer-ProgramsBrissan Guardado
 
AGNP or FNP: How to Choose?
AGNP or FNP: How to Choose?AGNP or FNP: How to Choose?
AGNP or FNP: How to Choose?smtibor
 
2016 Best of ASCO Brochure
2016 Best of ASCO Brochure2016 Best of ASCO Brochure
2016 Best of ASCO Brochureflasco_org
 

Was ist angesagt? (19)

Sandberg AAMC Innovations Report
Sandberg AAMC Innovations ReportSandberg AAMC Innovations Report
Sandberg AAMC Innovations Report
 
Murase2014
Murase2014Murase2014
Murase2014
 
Medical Students Who Want to Become PCPs Are Reconsidering Their Options
Medical Students Who Want to Become PCPs Are Reconsidering Their OptionsMedical Students Who Want to Become PCPs Are Reconsidering Their Options
Medical Students Who Want to Become PCPs Are Reconsidering Their Options
 
Best Practices: Needs Assessment Process, Data/Metric Tracking, and Survivors...
Best Practices: Needs Assessment Process, Data/Metric Tracking, and Survivors...Best Practices: Needs Assessment Process, Data/Metric Tracking, and Survivors...
Best Practices: Needs Assessment Process, Data/Metric Tracking, and Survivors...
 
Survivorship Program Examples
Survivorship Program ExamplesSurvivorship Program Examples
Survivorship Program Examples
 
Setting Up for Survivorship Success
Setting Up for Survivorship SuccessSetting Up for Survivorship Success
Setting Up for Survivorship Success
 
Medicine grand rounds
Medicine grand roundsMedicine grand rounds
Medicine grand rounds
 
Donna Hewitt Resume 2015
Donna Hewitt Resume 2015Donna Hewitt Resume 2015
Donna Hewitt Resume 2015
 
Pancreatic-Effective-Practices-in-Pancreatic-Cancer-Programs
Pancreatic-Effective-Practices-in-Pancreatic-Cancer-ProgramsPancreatic-Effective-Practices-in-Pancreatic-Cancer-Programs
Pancreatic-Effective-Practices-in-Pancreatic-Cancer-Programs
 
Managing a Navigation Program: The Role of Administrators
Managing a Navigation Program: The Role of AdministratorsManaging a Navigation Program: The Role of Administrators
Managing a Navigation Program: The Role of Administrators
 
Pre-Conference: The Navigator Matrix
Pre-Conference: The Navigator MatrixPre-Conference: The Navigator Matrix
Pre-Conference: The Navigator Matrix
 
Navigator_Jessica
Navigator_JessicaNavigator_Jessica
Navigator_Jessica
 
Patient Navigation Across the Continuum
Patient Navigation Across the ContinuumPatient Navigation Across the Continuum
Patient Navigation Across the Continuum
 
AGNP or FNP: How to Choose?
AGNP or FNP: How to Choose?AGNP or FNP: How to Choose?
AGNP or FNP: How to Choose?
 
ph capstone poster
ph capstone posterph capstone poster
ph capstone poster
 
2016 Best of ASCO Brochure
2016 Best of ASCO Brochure2016 Best of ASCO Brochure
2016 Best of ASCO Brochure
 
Primary Care in Paradise Brochure 2015
Primary Care in Paradise Brochure 2015Primary Care in Paradise Brochure 2015
Primary Care in Paradise Brochure 2015
 
AML NCCN 2017
AML NCCN 2017AML NCCN 2017
AML NCCN 2017
 
Removing Barriers to APRN Practice and Care: The Consumer Perspective
Removing Barriers to APRN Practice and Care: The Consumer PerspectiveRemoving Barriers to APRN Practice and Care: The Consumer Perspective
Removing Barriers to APRN Practice and Care: The Consumer Perspective
 

Ähnlich wie Cancer renal

Cistitis insterticial
Cistitis insterticialCistitis insterticial
Cistitis insterticial23762376
 
Incontinencia orina esfuerzo
Incontinencia orina esfuerzoIncontinencia orina esfuerzo
Incontinencia orina esfuerzo23762376
 
Eco prostatica
Eco prostaticaEco prostatica
Eco prostatica23762376
 
2010 Oncology Nursing Society Congress
2010 Oncology Nursing Society Congress2010 Oncology Nursing Society Congress
2010 Oncology Nursing Society CongressClinica de imagenes
 
Vejiga neurogénica
Vejiga neurogénicaVejiga neurogénica
Vejiga neurogénica23762376
 
MCI brochure 2005
MCI brochure 2005MCI brochure 2005
MCI brochure 2005wef
 
Imaging Techniques for the Diagnosis and Staging of Hepatocellular Carcinoma
Imaging Techniques for the Diagnosis and Staging of Hepatocellular CarcinomaImaging Techniques for the Diagnosis and Staging of Hepatocellular Carcinoma
Imaging Techniques for the Diagnosis and Staging of Hepatocellular CarcinomaImran Javed
 
Pathology-2016_Brochure
Pathology-2016_BrochurePathology-2016_Brochure
Pathology-2016_BrochureRebecca Paul
 
Clinician Webinar C E Flyer6 16 11
Clinician Webinar  C E  Flyer6 16 11Clinician Webinar  C E  Flyer6 16 11
Clinician Webinar C E Flyer6 16 11Julie Martinelli
 
MCI brochure 2008
MCI brochure 2008MCI brochure 2008
MCI brochure 2008wef
 
NMA Presentation Ddey 04Jun08
NMA Presentation Ddey 04Jun08NMA Presentation Ddey 04Jun08
NMA Presentation Ddey 04Jun08aplace2relax
 
MCI brochure2009
MCI brochure2009MCI brochure2009
MCI brochure2009wef
 
MCI brochure2010
MCI brochure2010MCI brochure2010
MCI brochure2010wef
 
Carlos Barrera Resume
Carlos Barrera Resume Carlos Barrera Resume
Carlos Barrera Resume Carlos Barrera
 
Diagnostic hemoglobinopathies Second Edition
Diagnostic hemoglobinopathies Second EditionDiagnostic hemoglobinopathies Second Edition
Diagnostic hemoglobinopathies Second Editionfssherwani
 
Olumi_CV_4_2016
Olumi_CV_4_2016Olumi_CV_4_2016
Olumi_CV_4_2016Aria Olumi
 
MCI brochure 2011
MCI brochure 2011MCI brochure 2011
MCI brochure 2011wef
 
Scripps 35th Annual Conference: Clinical Hematology & Oncology Brochure 2015
Scripps 35th Annual Conference: Clinical Hematology & Oncology Brochure 2015Scripps 35th Annual Conference: Clinical Hematology & Oncology Brochure 2015
Scripps 35th Annual Conference: Clinical Hematology & Oncology Brochure 2015Scripps Conference Services and CME
 
Anabolic Steroids
Anabolic SteroidsAnabolic Steroids
Anabolic SteroidsAmber Ford
 

Ähnlich wie Cancer renal (20)

Cistitis insterticial
Cistitis insterticialCistitis insterticial
Cistitis insterticial
 
Incontinencia orina esfuerzo
Incontinencia orina esfuerzoIncontinencia orina esfuerzo
Incontinencia orina esfuerzo
 
Eco prostatica
Eco prostaticaEco prostatica
Eco prostatica
 
2010 Oncology Nursing Society Congress
2010 Oncology Nursing Society Congress2010 Oncology Nursing Society Congress
2010 Oncology Nursing Society Congress
 
Vejiga neurogénica
Vejiga neurogénicaVejiga neurogénica
Vejiga neurogénica
 
MCI brochure 2005
MCI brochure 2005MCI brochure 2005
MCI brochure 2005
 
2015 CIB
2015 CIB2015 CIB
2015 CIB
 
Imaging Techniques for the Diagnosis and Staging of Hepatocellular Carcinoma
Imaging Techniques for the Diagnosis and Staging of Hepatocellular CarcinomaImaging Techniques for the Diagnosis and Staging of Hepatocellular Carcinoma
Imaging Techniques for the Diagnosis and Staging of Hepatocellular Carcinoma
 
Pathology-2016_Brochure
Pathology-2016_BrochurePathology-2016_Brochure
Pathology-2016_Brochure
 
Clinician Webinar C E Flyer6 16 11
Clinician Webinar  C E  Flyer6 16 11Clinician Webinar  C E  Flyer6 16 11
Clinician Webinar C E Flyer6 16 11
 
MCI brochure 2008
MCI brochure 2008MCI brochure 2008
MCI brochure 2008
 
NMA Presentation Ddey 04Jun08
NMA Presentation Ddey 04Jun08NMA Presentation Ddey 04Jun08
NMA Presentation Ddey 04Jun08
 
MCI brochure2009
MCI brochure2009MCI brochure2009
MCI brochure2009
 
MCI brochure2010
MCI brochure2010MCI brochure2010
MCI brochure2010
 
Carlos Barrera Resume
Carlos Barrera Resume Carlos Barrera Resume
Carlos Barrera Resume
 
Diagnostic hemoglobinopathies Second Edition
Diagnostic hemoglobinopathies Second EditionDiagnostic hemoglobinopathies Second Edition
Diagnostic hemoglobinopathies Second Edition
 
Olumi_CV_4_2016
Olumi_CV_4_2016Olumi_CV_4_2016
Olumi_CV_4_2016
 
MCI brochure 2011
MCI brochure 2011MCI brochure 2011
MCI brochure 2011
 
Scripps 35th Annual Conference: Clinical Hematology & Oncology Brochure 2015
Scripps 35th Annual Conference: Clinical Hematology & Oncology Brochure 2015Scripps 35th Annual Conference: Clinical Hematology & Oncology Brochure 2015
Scripps 35th Annual Conference: Clinical Hematology & Oncology Brochure 2015
 
Anabolic Steroids
Anabolic SteroidsAnabolic Steroids
Anabolic Steroids
 

Mehr von 23762376

Aua cáncer renal
Aua cáncer renalAua cáncer renal
Aua cáncer renal23762376
 
Aua manejo y diagnostico calculo ureteral
Aua manejo y diagnostico calculo ureteralAua manejo y diagnostico calculo ureteral
Aua manejo y diagnostico calculo ureteral23762376
 
Aua manejo y diagnostico calculo ureteral
Aua manejo y diagnostico calculo ureteralAua manejo y diagnostico calculo ureteral
Aua manejo y diagnostico calculo ureteral23762376
 
Cancer renal
Cancer renalCancer renal
Cancer renal23762376
 
Caso clinico
Caso clinicoCaso clinico
Caso clinico23762376
 
Reseccionprost adenoma
Reseccionprost adenomaReseccionprost adenoma
Reseccionprost adenoma23762376
 
Reseccion transuret
Reseccion transuretReseccion transuret
Reseccion transuret23762376
 
Prostt radical
Prostt radicalProstt radical
Prostt radical23762376
 
Nefrect ampliada
Nefrect ampliadaNefrect ampliada
Nefrect ampliada23762376
 
Litotricia ec
Litotricia ecLitotricia ec
Litotricia ec23762376
 
Cistoscopia
CistoscopiaCistoscopia
Cistoscopia23762376
 
Circuncision
CircuncisionCircuncision
Circuncision23762376
 
04 hidrocele
04 hidrocele04 hidrocele
04 hidrocele23762376
 
02 biopsias prostaticas
02 biopsias prostaticas02 biopsias prostaticas
02 biopsias prostaticas23762376
 
01 adenomectomia
01 adenomectomia01 adenomectomia
01 adenomectomia23762376
 
Los tumores testiculares
Los tumores testicularesLos tumores testiculares
Los tumores testiculares23762376
 
Tuberculosis urinaria
Tuberculosis urinariaTuberculosis urinaria
Tuberculosis urinaria23762376
 
Clase de cáncer de próstata
Clase de cáncer de próstataClase de cáncer de próstata
Clase de cáncer de próstata23762376
 
Litiasis renal
Litiasis renal Litiasis renal
Litiasis renal 23762376
 
Varicocele y fertilidad
Varicocele y fertilidadVaricocele y fertilidad
Varicocele y fertilidad23762376
 

Mehr von 23762376 (20)

Aua cáncer renal
Aua cáncer renalAua cáncer renal
Aua cáncer renal
 
Aua manejo y diagnostico calculo ureteral
Aua manejo y diagnostico calculo ureteralAua manejo y diagnostico calculo ureteral
Aua manejo y diagnostico calculo ureteral
 
Aua manejo y diagnostico calculo ureteral
Aua manejo y diagnostico calculo ureteralAua manejo y diagnostico calculo ureteral
Aua manejo y diagnostico calculo ureteral
 
Cancer renal
Cancer renalCancer renal
Cancer renal
 
Caso clinico
Caso clinicoCaso clinico
Caso clinico
 
Reseccionprost adenoma
Reseccionprost adenomaReseccionprost adenoma
Reseccionprost adenoma
 
Reseccion transuret
Reseccion transuretReseccion transuret
Reseccion transuret
 
Prostt radical
Prostt radicalProstt radical
Prostt radical
 
Nefrect ampliada
Nefrect ampliadaNefrect ampliada
Nefrect ampliada
 
Litotricia ec
Litotricia ecLitotricia ec
Litotricia ec
 
Cistoscopia
CistoscopiaCistoscopia
Cistoscopia
 
Circuncision
CircuncisionCircuncision
Circuncision
 
04 hidrocele
04 hidrocele04 hidrocele
04 hidrocele
 
02 biopsias prostaticas
02 biopsias prostaticas02 biopsias prostaticas
02 biopsias prostaticas
 
01 adenomectomia
01 adenomectomia01 adenomectomia
01 adenomectomia
 
Los tumores testiculares
Los tumores testicularesLos tumores testiculares
Los tumores testiculares
 
Tuberculosis urinaria
Tuberculosis urinariaTuberculosis urinaria
Tuberculosis urinaria
 
Clase de cáncer de próstata
Clase de cáncer de próstataClase de cáncer de próstata
Clase de cáncer de próstata
 
Litiasis renal
Litiasis renal Litiasis renal
Litiasis renal
 
Varicocele y fertilidad
Varicocele y fertilidadVaricocele y fertilidad
Varicocele y fertilidad
 

Kürzlich hochgeladen

Book Call Girls in Yelahanka - For 7001305949 Cheap & Best with original Photos
Book Call Girls in Yelahanka - For 7001305949 Cheap & Best with original PhotosBook Call Girls in Yelahanka - For 7001305949 Cheap & Best with original Photos
Book Call Girls in Yelahanka - For 7001305949 Cheap & Best with original Photosnarwatsonia7
 
Sonagachi Call Girls Services 9907093804 @24x7 High Class Babes Here Call Now
Sonagachi Call Girls Services 9907093804 @24x7 High Class Babes Here Call NowSonagachi Call Girls Services 9907093804 @24x7 High Class Babes Here Call Now
Sonagachi Call Girls Services 9907093804 @24x7 High Class Babes Here Call NowRiya Pathan
 
Housewife Call Girls Bangalore - Call 7001305949 Rs-3500 with A/C Room Cash o...
Housewife Call Girls Bangalore - Call 7001305949 Rs-3500 with A/C Room Cash o...Housewife Call Girls Bangalore - Call 7001305949 Rs-3500 with A/C Room Cash o...
Housewife Call Girls Bangalore - Call 7001305949 Rs-3500 with A/C Room Cash o...narwatsonia7
 
Russian Call Girl Brookfield - 7001305949 Escorts Service 50% Off with Cash O...
Russian Call Girl Brookfield - 7001305949 Escorts Service 50% Off with Cash O...Russian Call Girl Brookfield - 7001305949 Escorts Service 50% Off with Cash O...
Russian Call Girl Brookfield - 7001305949 Escorts Service 50% Off with Cash O...narwatsonia7
 
call girls in green park DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️
call girls in green park  DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️call girls in green park  DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️
call girls in green park DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️saminamagar
 
Aspirin presentation slides by Dr. Rewas Ali
Aspirin presentation slides by Dr. Rewas AliAspirin presentation slides by Dr. Rewas Ali
Aspirin presentation slides by Dr. Rewas AliRewAs ALI
 
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.Artifacts in Nuclear Medicine with Identifying and resolving artifacts.
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.MiadAlsulami
 
Ahmedabad Call Girls CG Road 🔝9907093804 Short 1500 💋 Night 6000
Ahmedabad Call Girls CG Road 🔝9907093804  Short 1500  💋 Night 6000Ahmedabad Call Girls CG Road 🔝9907093804  Short 1500  💋 Night 6000
Ahmedabad Call Girls CG Road 🔝9907093804 Short 1500 💋 Night 6000aliya bhat
 
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy Girls
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy GirlsCall Girls In Andheri East Call 9920874524 Book Hot And Sexy Girls
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy Girlsnehamumbai
 
College Call Girls Vyasarpadi Whatsapp 7001305949 Independent Escort Service
College Call Girls Vyasarpadi Whatsapp 7001305949 Independent Escort ServiceCollege Call Girls Vyasarpadi Whatsapp 7001305949 Independent Escort Service
College Call Girls Vyasarpadi Whatsapp 7001305949 Independent Escort ServiceNehru place Escorts
 
Call Girls Frazer Town Just Call 7001305949 Top Class Call Girl Service Avail...
Call Girls Frazer Town Just Call 7001305949 Top Class Call Girl Service Avail...Call Girls Frazer Town Just Call 7001305949 Top Class Call Girl Service Avail...
Call Girls Frazer Town Just Call 7001305949 Top Class Call Girl Service Avail...narwatsonia7
 
Mumbai Call Girls Service 9910780858 Real Russian Girls Looking Models
Mumbai Call Girls Service 9910780858 Real Russian Girls Looking ModelsMumbai Call Girls Service 9910780858 Real Russian Girls Looking Models
Mumbai Call Girls Service 9910780858 Real Russian Girls Looking Modelssonalikaur4
 
Call Girls Hebbal Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Hebbal Just Call 7001305949 Top Class Call Girl Service AvailableCall Girls Hebbal Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Hebbal Just Call 7001305949 Top Class Call Girl Service Availablenarwatsonia7
 
Call Girls Service Chennai Jiya 7001305949 Independent Escort Service Chennai
Call Girls Service Chennai Jiya 7001305949 Independent Escort Service ChennaiCall Girls Service Chennai Jiya 7001305949 Independent Escort Service Chennai
Call Girls Service Chennai Jiya 7001305949 Independent Escort Service ChennaiNehru place Escorts
 
Call Girls Hosur Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Hosur Just Call 7001305949 Top Class Call Girl Service AvailableCall Girls Hosur Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Hosur Just Call 7001305949 Top Class Call Girl Service Availablenarwatsonia7
 
Book Call Girls in Kasavanahalli - 7001305949 with real photos and phone numbers
Book Call Girls in Kasavanahalli - 7001305949 with real photos and phone numbersBook Call Girls in Kasavanahalli - 7001305949 with real photos and phone numbers
Book Call Girls in Kasavanahalli - 7001305949 with real photos and phone numbersnarwatsonia7
 
Russian Call Girls in Pune Riya 9907093804 Short 1500 Night 6000 Best call gi...
Russian Call Girls in Pune Riya 9907093804 Short 1500 Night 6000 Best call gi...Russian Call Girls in Pune Riya 9907093804 Short 1500 Night 6000 Best call gi...
Russian Call Girls in Pune Riya 9907093804 Short 1500 Night 6000 Best call gi...Miss joya
 
Call Girls Jp Nagar Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Jp Nagar Just Call 7001305949 Top Class Call Girl Service AvailableCall Girls Jp Nagar Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Jp Nagar Just Call 7001305949 Top Class Call Girl Service Availablenarwatsonia7
 
Call Girls Service in Bommanahalli - 7001305949 with real photos and phone nu...
Call Girls Service in Bommanahalli - 7001305949 with real photos and phone nu...Call Girls Service in Bommanahalli - 7001305949 with real photos and phone nu...
Call Girls Service in Bommanahalli - 7001305949 with real photos and phone nu...narwatsonia7
 
Asthma Review - GINA guidelines summary 2024
Asthma Review - GINA guidelines summary 2024Asthma Review - GINA guidelines summary 2024
Asthma Review - GINA guidelines summary 2024Gabriel Guevara MD
 

Kürzlich hochgeladen (20)

Book Call Girls in Yelahanka - For 7001305949 Cheap & Best with original Photos
Book Call Girls in Yelahanka - For 7001305949 Cheap & Best with original PhotosBook Call Girls in Yelahanka - For 7001305949 Cheap & Best with original Photos
Book Call Girls in Yelahanka - For 7001305949 Cheap & Best with original Photos
 
Sonagachi Call Girls Services 9907093804 @24x7 High Class Babes Here Call Now
Sonagachi Call Girls Services 9907093804 @24x7 High Class Babes Here Call NowSonagachi Call Girls Services 9907093804 @24x7 High Class Babes Here Call Now
Sonagachi Call Girls Services 9907093804 @24x7 High Class Babes Here Call Now
 
Housewife Call Girls Bangalore - Call 7001305949 Rs-3500 with A/C Room Cash o...
Housewife Call Girls Bangalore - Call 7001305949 Rs-3500 with A/C Room Cash o...Housewife Call Girls Bangalore - Call 7001305949 Rs-3500 with A/C Room Cash o...
Housewife Call Girls Bangalore - Call 7001305949 Rs-3500 with A/C Room Cash o...
 
Russian Call Girl Brookfield - 7001305949 Escorts Service 50% Off with Cash O...
Russian Call Girl Brookfield - 7001305949 Escorts Service 50% Off with Cash O...Russian Call Girl Brookfield - 7001305949 Escorts Service 50% Off with Cash O...
Russian Call Girl Brookfield - 7001305949 Escorts Service 50% Off with Cash O...
 
call girls in green park DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️
call girls in green park  DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️call girls in green park  DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️
call girls in green park DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️
 
Aspirin presentation slides by Dr. Rewas Ali
Aspirin presentation slides by Dr. Rewas AliAspirin presentation slides by Dr. Rewas Ali
Aspirin presentation slides by Dr. Rewas Ali
 
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.Artifacts in Nuclear Medicine with Identifying and resolving artifacts.
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.
 
Ahmedabad Call Girls CG Road 🔝9907093804 Short 1500 💋 Night 6000
Ahmedabad Call Girls CG Road 🔝9907093804  Short 1500  💋 Night 6000Ahmedabad Call Girls CG Road 🔝9907093804  Short 1500  💋 Night 6000
Ahmedabad Call Girls CG Road 🔝9907093804 Short 1500 💋 Night 6000
 
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy Girls
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy GirlsCall Girls In Andheri East Call 9920874524 Book Hot And Sexy Girls
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy Girls
 
College Call Girls Vyasarpadi Whatsapp 7001305949 Independent Escort Service
College Call Girls Vyasarpadi Whatsapp 7001305949 Independent Escort ServiceCollege Call Girls Vyasarpadi Whatsapp 7001305949 Independent Escort Service
College Call Girls Vyasarpadi Whatsapp 7001305949 Independent Escort Service
 
Call Girls Frazer Town Just Call 7001305949 Top Class Call Girl Service Avail...
Call Girls Frazer Town Just Call 7001305949 Top Class Call Girl Service Avail...Call Girls Frazer Town Just Call 7001305949 Top Class Call Girl Service Avail...
Call Girls Frazer Town Just Call 7001305949 Top Class Call Girl Service Avail...
 
Mumbai Call Girls Service 9910780858 Real Russian Girls Looking Models
Mumbai Call Girls Service 9910780858 Real Russian Girls Looking ModelsMumbai Call Girls Service 9910780858 Real Russian Girls Looking Models
Mumbai Call Girls Service 9910780858 Real Russian Girls Looking Models
 
Call Girls Hebbal Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Hebbal Just Call 7001305949 Top Class Call Girl Service AvailableCall Girls Hebbal Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Hebbal Just Call 7001305949 Top Class Call Girl Service Available
 
Call Girls Service Chennai Jiya 7001305949 Independent Escort Service Chennai
Call Girls Service Chennai Jiya 7001305949 Independent Escort Service ChennaiCall Girls Service Chennai Jiya 7001305949 Independent Escort Service Chennai
Call Girls Service Chennai Jiya 7001305949 Independent Escort Service Chennai
 
Call Girls Hosur Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Hosur Just Call 7001305949 Top Class Call Girl Service AvailableCall Girls Hosur Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Hosur Just Call 7001305949 Top Class Call Girl Service Available
 
Book Call Girls in Kasavanahalli - 7001305949 with real photos and phone numbers
Book Call Girls in Kasavanahalli - 7001305949 with real photos and phone numbersBook Call Girls in Kasavanahalli - 7001305949 with real photos and phone numbers
Book Call Girls in Kasavanahalli - 7001305949 with real photos and phone numbers
 
Russian Call Girls in Pune Riya 9907093804 Short 1500 Night 6000 Best call gi...
Russian Call Girls in Pune Riya 9907093804 Short 1500 Night 6000 Best call gi...Russian Call Girls in Pune Riya 9907093804 Short 1500 Night 6000 Best call gi...
Russian Call Girls in Pune Riya 9907093804 Short 1500 Night 6000 Best call gi...
 
Call Girls Jp Nagar Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Jp Nagar Just Call 7001305949 Top Class Call Girl Service AvailableCall Girls Jp Nagar Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Jp Nagar Just Call 7001305949 Top Class Call Girl Service Available
 
Call Girls Service in Bommanahalli - 7001305949 with real photos and phone nu...
Call Girls Service in Bommanahalli - 7001305949 with real photos and phone nu...Call Girls Service in Bommanahalli - 7001305949 with real photos and phone nu...
Call Girls Service in Bommanahalli - 7001305949 with real photos and phone nu...
 
Asthma Review - GINA guidelines summary 2024
Asthma Review - GINA guidelines summary 2024Asthma Review - GINA guidelines summary 2024
Asthma Review - GINA guidelines summary 2024
 

Cancer renal

  • 1. Kidney Cancer: Recent Advances in Diagnosis and Management Saturday, May 17, 2008 6:00- 8:00 p.m. COURSE 08 EC FACULTY W. Marston Linehan, M.D., Ph.D. Course Director Peter A. Pinto, M.D. Jeffrey A. Sosman, M.D. American Urological Association Education and Research Inc. 2008 Annual Meeting, Orlando, FL May 17-22, 2008 Sponsored by: The American Urological Association Education and Research, Inc.
  • 2. Meeting Disclaimer Regarding materials and information received, written or otherwise, during the 2008 American Urological Association Education and Research, Inc. Annual Meeting Instructional/Postgraduate MC/EC and Dry Lab Courses sponsored by the Office of Education: The scientific views, statements, and recommendations expressed in the written materials and during the meeting represent those of the authors and speakers and do not necessarily represent the views of the American Urological Association Education and Research, Inc.® Reproduction Permission Reproduction of all Instructional/Postgraduate, MC/EC and Dry Lab Courses is prohibited without written permission from individual authors and the American Urological Association Education and Research, Inc. These materials have been written and produced as a supplement to continuing medical education activities pursued during the Instructional/Postgraduate, MC/EC and Dry Lab Courses and are intended for use in that context only. Use of this material as an educational tool or singular resource/authority on the subject/s outside the context of the meeting is not intended. Accreditation The American Urological Association Education and Research, Inc. is accredited by the Accreditation Council for Continuing Medical Education (ACCME) to provide continuing medical education (CME) for physicians. The American Urological Association Education and Research, Inc. takes responsibility for the content, quality, and scientific integrity of the CME activity. CME Credit The American Urological Association Education and Research, Inc. designates each Instructional Course educational activity for a maximum of 1.5 AMA PRA Category 1 credits™; each Postgraduate Course for a maximum of 3.25 AMA PRA Category 1 credits™; each MC Course for a maximum of 1.0 AMA PRA Category 1 credits™; each EC Course for a maximum of 2.0 AMA PRA Category 1 credits™; each MC Plus Course for a maximum of 2.0 AMA PRA Category 1 credits™; and each Dry Lab Course for a maximum of 2.5 AMA PRA Category 1 credits™. Physicians should only claim credits commensurate with the extent of their participation in the activity. Disclosure Policy Statement As a provider accredited by the Accreditation Council for Continuing Medical Education (ACCME), the American Urological Association Education and Research, Inc., must insure balance, independence, objectivity and scientific rigor in all its sponsored activities. All faculty participating in an educational activity provided by the American Urological Association Education and Research, Inc. are required to disclose to the audience any relevant financial relationships with any commercial interest to the provider. The intent of this disclosure is not to prevent a faculty with relevant financial relationships from serving as faculty, but rather to provide members of the audience with information on which they can make their own judgments. The American Urological Association Education and Research, Inc. must resolve any conflicts of interest prior to the commencement of the educational activity. It remains for the audience to determine if the faculty’s relationships may influence the educational content with regard to exposition or conclusion. When unlabeled or unapproved uses are discussed, these are also indicated. Evidence-based Content As a provider of continuing medical education accredited by the Accreditation Council for Continuing Medical Education (ACCME), it is the policy of the American Urological Association Education and Research, Inc. to review and certify that the content contained in this CME activity is evidence-based, valid, fair and balanced, scientifically rigorous, and free of commercial bias.
  • 3. 2008 AUA Annual Meeting 08 EC Kidney Cancer: Recent Advances in Diagnosis and Management 5/17/2008 6:00 - 8:00 p.m. Disclosures According to the American Urological Association’s Disclosure Policy, speakers involved in continuing medical education activities are required to report all relevant financial relationships with any commercial interest to the provider by completing an AUA Disclosure Form. All information from this form is provided to meeting participants so that they may make their own judgments about a speaker’s presentation. Well in advance of the CME activity, all disclosure information is reviewed by a peer group for identification of conflicts of interest, which are resolved in a variety of ways. The American Urological Association does not view the existence of relevant financial relationships as necessarily implying bias, conflict of interest, or decreasing the value of the presentation. Each faculty member presenting lectures in the Annual Meeting Instructional or Postgraduate, MC or EC and Dry Lab Courses has submitted a copy of his or her Disclosure online to the AUA. These copies are on file in the AUA Office of Education. This course has been planned to be well balanced, objective, and scientifically rigorous. Information and opinions offered by the speakers represent their viewpoints. Conclusions drawn by the audience members should be derived from careful consideration of all available scientific information. The following faculty members(s) declare a relationship with the commercial interests as listed below, related directly or indirectly to this CME activity. Participants may form their own judgments about the presentations in light of full disclosure of the facts. Faculty Disclosure W. Marston Linehan, M.D., Ph.D. Course Director National Cancer Institute: Meeting Participant or Lecturer Peter A. Pinto, M.D. Nothing to disclose Jeffrey A. Sosman, M.D. Nothing to disclose
  • 4. Disclosure of Off-Label Uses The audience is advised that this continuing medical education activity may contain reference(s) to unlabeled or unapproved uses of drugs or devices. Please consult the prescribing information for full disclosure of approved uses. Faculty and speakers are required to disclose unlabeled or unapproved use of drugs or devices before their presentation or discussion during this activity. A special AUA value for your patients: www.UrologyHealth.org is a joint AUA/AFUD patient education web site that provides accurate and unbiased information on urologic disease and conditions. It also provides information for patients and others wishing to locate urologists in their local areas. This site does not provide medical advice. The content and illustrations are for informational purposes only. This information is not intended to substitute for a consultation with a urologist. It is offered to educate the patient, and their families, in order for them to get the most out of office visits and consultations.
  • 5. Commercial Support Acknowledgements AUA Office of Education thanks the companies and representatives who support continuing education of physicians with unrestricted educational grants, displays at educational meetings, and their expertise. This is a valuable resource to everyone. The AUA Office of Education salutes the following company for providing an educational grant in support of the Kidney Cancer: Recent Advances in Diagnosis and Management course at the 2008 Annual Meeting. Wyeth Pharmaceuticals The scientific programs and faculty are independently developed and finalized by the Chair of Education, the Director of Education and Scientific Program Director with no influence or input from these or any companies providing either an unrestricted educational grant or technical support. Comments or letters regarding this program and/or company involvement in an educational meeting may be addressed to: Jan Baum, M.A. Director of Education American Urological Association 1000 Corporate Boulevard Linthicum, MD 21090 (410) 689-3930
  • 6.
  • 7. Recent Advances in Diagnosis and Management of Kidney Cancer W. Marston Linehan, M.D. Chief, Urologic Oncology Branch National Cancer Institute Bldg 10 Rm 1-5942 Bethesda, Maryland 20892-1107 Tel: (301) 496-6353 Fax: (301) 402-0922 Email: Email: WML@nih.gov Peter Pinto, M.D. Senior Investigator, Urologic Oncology Branch National Cancer Institute Bldg 10 Rm 2-5940 Bethesda, Maryland 20892-1107 Tel: (301) 496-6353 Fax: (301) 402-0922 Email : pintop@mail.nih.gov Jeffrey A. Sosman, M.D. Professor of Medicine (Hematology/Oncology) Vanderbilt-Ingram Cancer 777 Preston Building Nashville, TN 27232-6307 Office (615) 343-7602 Email: jeff.sosman@Vanderbilt.Edu
  • 8. RECENT ADVANCES IN DIAGNOSES AND MANAGEMENT OF KIDNEY CANCER Saturday, May 17, 2008 6:00PM - 8:00PM From To Speaker Topic 6:00 PM - 6:20 PM W. Marston Linehan, M.D. I. Molecular Genetics of Renal Carcinoma: Implications for the Urologic Surgeon II. Hereditary Forms of Renal Cancers 6:20 PM - 7:00 PM Peter A. Pinto, M.D. III. Techniques and Indications A) Laparoscopic Nephrectomy B) Laparoscopic Partial Nephrectomy IV. Robotic Assisted Nephrectomy and Partial Nephrectomy 7:00 PM - 7:40 PM Jeffrey A. Sosman, M.D. V. Role of Targeted Systemic Therapy In the Management of Advanced Clear Cell And Non-Clear Cell Kidney Cancer A) Sorafenib B) Sunitinib C) Bevacizumab D) Temsirolimus 7:40 PM - 8:00 PM W. Marston Linehan, M.D. VI. A) Management of Patients Who Present with Peter A. Pinto, M.D. Advanced Disease with Kidney In Place Jeffrey A. Sosman, M.D. B) Role of Adjuvant Therapy in the Management of Patients with High Risk Kidney Cancer 2
  • 9. I. Introduction Kidney cancer affects over 39,000 Americans annually and it is estimated that nearly 13,000 will die of this disease in the United States each year. Patients who are found to have the disease when it is localized to the kidney can have up to a 95% 5 and 10 year survival rate. Patients who present to their urologic surgeon with locally advanced disease have up to a 65% chance of surviving 5 years. Those who present with advanced disease have an 18% 2 year survival rate.1,2 The advent of laparoscopic surgery has provided new and significantly less morbid surgical strategies for the management of patients with localized, locally advanced and even advanced renal carcinoma. There have been exciting advances in the development of new minimally invasive strategies for the management of patients with localized kidney cancer involving radio frequency ablation therapy as well as cryotherapy. Recent advances in immunologic and molecular targeted forms of therapy have provided new hope for patients with advanced renal cell carcinoma.3,4 1. Molecular Genetics of Renal Carcinoma: Implications for the Urologic Surgeon (W. Marston Linehan, M.D.) Kidney cancer is not a single disease, it is made up of a number of different types of cancer that occur in the kidney; each with a different histology, having a different clinical course, responding differently to therapy and caused by different genes. Figure 1 Kidney cancer is made up of a number of different types of cancer, each with a different histology, with a different clinical course, responding differently to therapy and caused by different genes. From Linehan, et al.5 This section of the course will update you on current studies of the molecular genetics of sporadic and familial renal cancer, background on kidney cancer tumor suppressor genes and oncogenes, data for why we conclude that the von Hippel Lindau disease gene and the Met genes are critical genes for both hereditary and non-familial, sporadic kidney cancers, for how these genes fit the definitions of cancer genes and how these types of findings could lead to better methods for early diagnosis, prevention and, potentially, therapy of renal cancers will be presented. 3
  • 10. 2. Tumor Suppressor Genes and Oncogenes A. Tumor Suppressor Genes The pioneering studies of Dr. Alfred Knudson led to the development of a hypothesis that the most basic aspect of cancer would involve what is called a tumor suppressor gene. Instead of cancer being the result of activation of a gene such as an oncogene, which drives the abnormal proliferation that we know of as cancer, Knudson hypothesized that, instead, cancer could be the result of inactivation of a gene, a so called tumor suppressor gene, whose normal function was to regulate or control cellular growth. We each have two copies of each gene, one maternal and the other paternal. If both copies of a tumor suppressor gene were inactivated (Knudson “two-hit” hypothesis) by either loss of DNA (DNA sequence deletion), or another mechanism such as mutation, transformation would result. Knudson hypothesized that if this were the case in a sporadic, non-hereditary cancer, and there were a hereditary variant of the same malignancy, that the same gene might be involved in the origin of both. This is the case in retinoblastoma, where both copies of the retinoblastoma gene are inactivated in both the sporadic and hereditary form of the disease. Abnormalities of tumor suppressor genes have been found in a number of solid human tumors, including breast, bladder and, as we shall see, kidney cancer. The VHL kidney cancer tumor suppressor gene, the gene for clear cell renal carcinoma, is a classic tumor suppressor gene located on chromosome 3. B. Oncogene An oncogene is a “gain of function gene”; i.e., a gene in which a mutation of the gene results in activation of the gene, resulting in the unregulated growth that we know of as cancer. Often an oncogene may be “duplicated” in a cancer, resulting in three copies of the gene (instead of two copies). This is called trisomy. The c-Met oncogene, the gene for Hereditary Papillary Renal Carcinoma (HPRC), is a classic oncogene located on chromosome 7. 4
  • 11. II. Hereditary Forms of Renal Cancers Kidney cancer is like retinoblastoma and colon cancer in that there is both a sporadic and a familial form. Familial kidney cancer is far more prevalent than had been previously imagined. It is estimated that 5% of kidney cancer is familial (hereditary). There are four types of hereditary kidney cancer. The well described is clear cell renal cancer associated with von Hippel Lindau (VHL); there is a hereditary form of type 1 papillary renal carcinoma (HPRC); the third type is hereditary chromophobe renal carcinoma associated with Birt Hogg Dubé (BHD) and the fourth is a hereditary type of papillary type 2 renal carcinoma associated with Hereditary Leiomyomatosis Renal Carcinoma (HLRCC). Table 1 Inherited Forms of Renal Carcinoma 1. Von Hippel Lindau (VHL) Clear Cell RCC 2. Hereditary Papillary Renal Carcinoma (HPRC) Papillary Type 1 RCC 3. Birt Hogg Dubé (BHD) Chromophobe RCC/Oncocytoma 4. Hereditary Leiomyomatosis RCC (HLRCC) Papillary Type 2 RCC These familial forms of kidney cancer are different from sporadic, non-familial kidney cancer in that they tend to be multifocal, bilateral and occur at a younger age, suggesting a genetic predisposition to develop these cancers. 1. Von Hippel Lindau (VHL) A. Renal Carcinoma Gene Localization Studies: Inherited Renal Cancer To identify the disease gene for clear cell kidney cancer the familial form of renal cell carcinoma associated with von Hippel Lindau (VHL) was studied. Von Hippel Lindau is an autosomal dominant hereditary cancer syndrome in which affected individuals are at risk to develop tumors in a number of organs, including the kidneys. VHL patients are at risk to develop multiple, bilateral renal carcinomas and cysts, cerebellar and spinal hemangioblastomas, pheochromocytomas, retinal angiomas, epididymal cystadenomas, pancreatic cysts and islet cell tumors and tumors in the inner ear, endolymphatic sac tumors. 5
  • 12. Table 2 Clinical Features of VHL h Kidney tumors (clear cell RCC) h Adrenal tumors (pheochromocytoma) h Pancreatic tumors (islet tumors) h CNS tumors (hemangioblastoma) h Retinal tumors (angioma) h Endolymphatic sac tumors (inner ear) h Epididymal cystadenoma B. VHL: Clinical Evaluation Clinical evaluation of VHL patients involves MRI of the brain and spine (for CNS hemangioblastomas), abdominal CT and ultrasound (for evaluation of renal, adrenal and pancreatic manifestations of VHL), ophthalmologic evaluation (for evaluation for retinal hemangiomas), audiometric and ENT evaluation (to search for presence of ELST tumors), testicular ultrasound (to evaluate epididymal cystadenomas). Table 3 Clinical Evaluation-VHL • VHL gene germline mutation testing • MRI of the brain and spine • Abdominal CT and ultrasound • Ophthalmologic evaluation • Audiometric and ENT evaluation • Testicular Ultrasound • Metabolic evaluation (catechols) C. VHL: Surgical Management Surgical management of the renal manifestations of VHL patients involves nephron sparing surgery whenever possible. Patients with small renal tumors, generally under 2.5 cm, are often managed with expectant management. When the tumors reach 3 cm, surgery is often recommended. As it has been estimated that there can be up to 600 tumors per kidney in VHL patients, surgical resection of renal lesions is not considered “curative.” Rather, it is considered that surgical management will hopefully “set back the clock”; i.e. help prevent metastasis. Historically 35 to 45% of VHL patients have died of complications of metastatic renal cell carcinoma. The decision to recommend surgery must balance the risk of metastasis with the morbidity of surgery. This is often a complex issue in VHL patients with multisystem manifestations. When surgery is performed, thorough evaluation of the kidney with intraoperative ultrasound is considered a valuable adjunct to the surgical procedure. This allows 6
  • 13. the surgeon to localize renal tumors and cysts and to perform as thorough and safe a procedure as possible. Renal Lesions in VHL Patient Microscopic VHL RCC Focus It is important that VHL patients undergo a complete screening and metabolic evaluation prior to a surgical procedure, to rule out such unsuspected manifestations as a CNS hemangioblastoma or pheochromocytoma. D. The VHL gene is on Chromosome 3 In order to identify the gene associated with VHL we evaluated affected and at risk individuals for VHL. DNA was extracted from blood of over 4,000 individuals. 450 patients were screened at the NIH for the presence of von Hippel Lindau. In order to localize this kidney cancer disease gene genetic linkage analysis was utilized, which narrowed the area of the VHL gene to a small region on chromosome 3, which is the same region where we showed abnormalities in tumor tissue from patients with non-familial, clear cell kidney cancer. Mutations of the VHL Gene in the Germline of VHL Patients From Chen et al.6 E. The VHL gene is the gene for sporadic, non-inherited clear cell RCC Mutations of the VHL gene is tumors from patients with sporadic, non- hereditary clear cell kidney cancer. From Gnarra, et al. 7 VHL mutations have been identified in a high percentage of tumors from patients with sporadic, clear cell renal carcinomas. VHL somatic mutations have been detected in the earliest, 7
  • 14. localized clear cell tumors. No mutations have been found in papillary, chromophobe, collecting duct or other types of renal tumors. F. Molecular Targeting the VHL gene in clear cell kidney cancer It is hoped that understanding the pathways for the genes that cause kidney cancer will lead to the development of more effective forms of therapy for patients with this disease. The VHL clear cell kidney cancer gene was identified in 1993 and since that time an intensive effort has been underway to identify how damage to this gene leads to kidney cancer. The VHL gene product has been found to be important regulator of Hypoxia Inducible Factor (HIF). HIF is a transcription factor which regulates a number of downstream genes known to be important in cancer, such as vascular endothelial growth factor (VEGF), platelet derived growth factor (PDGF), erythropoietin, etc. Scientists are currently evaluating the role of small molecules that block this pathway as potential strategies for therapy. VHL Kidney Cancer Gene Pathway RCC Molecular targeting: anti-VEGF antibody. Adapted from Linehan, et al.5 The FDA recently approved three new agents, sunitinib, sorafenib and temsirolimus for the treatment of patients with advanced kidney cancer. Targeting VHL/HIF in Clear Cell RCC VHL HIF VEGF PDGF TGF-α VEGFR PDGFR EGFR Sunitinib Sunitinib The partial response rate in patients with advanced renal carcinoma treated with sunitinib has been reported to be 31% and there was an increase in progression free survival in patients treated with sunitinib versus interferon.8 The most commonly reported side effects with sunitinib involve diarrhea, skin discoloration, mouth irritation, weakness, and altered taste. Patients treated with sunitinib may also experience, high blood pressure, fatigue, bleeding, and swelling. 8
  • 15. Targeting VHL/HIF in Clear Cell RCC VHL HIF VEGF PDGF TGF-α VEGFR PDGFR EGFR Raf Raf Sorafenib Sorafenib Sorafenib Sorafenib The partial response rate in patients with advanced renal carcinoma treated with sorafenib has been reported to be 10% and the progression free survival was 5.5 months versus 2.8 months for patients treated with placebo.9 Targeting VHL/HIF in Clear Cell RCC VHL HIF mTOR Temsirolimus VEGF PDGF TGF-α VEGFR PDGFR EGFR Temsirolimus (inhibitor of mammalian target of rapamycin-MTOR) Temsirolimus, the MTOR-inhibiting agent, has been found to have activity in patients with advanced, poor prognosis kidney cancer. In a recent report of a randomized three-arm phase 3 trial, temsirolimus was found to have an overall statistically significant survival benefit versus treatment with interferon alone (10.9 versus 7.3 months). This study confirmed MTOR as a valid target in kidney cancer and that temsirolimus monotherapy prolongs survival in patients with poor prognosis kidney cancer. 10 G. Adjuvant Treatment The role of agents such as sunitinib and sorafenib in the adjuvant setting are not known. To address this question, a randomized phase 3 trial involving 1332 patients, supported by the Eastern Cooperative Oncology Group (ECOG), the Cancer Trials Support Unit CTSU), the Southwest Oncology Group (SWOG), the Cancer and Leukemia Group B (CALGB) and the National Cancer Institute of Canada (NCIC). In this trial, patients with T1B, T2 or T4 (N0-N2) will be randomized to receive one year of sunitinib, sorafenib or placebo after nephrectomy.11 Information about the trial can be found at the Kidney Cancer Association web site. 9
  • 16. 2. Hereditary Papillary Renal Cell Carcinoma: HPRC A. HPRC: Clinical Features Hereditary Papillary Renal Carcinoma is a hereditary cancer syndrome in which affected individuals are at risk for the development of bilateral, multifocal, type 1 papillary renal carcinoma.12 HPRC patients are at risk for the development of up to 3,000 tumors per kidney, which are uniformly of type 1 papillary histologic type.13 Figure 2 Hereditary Papillary Renal Carcinoma (HPRC) is a hereditary cancer syndrome where affected individuals are at risk for the development of bilateral, multifocal, type 1 papillary renal carcinoma. From Linehan, et al.5 Table 4 Hereditary Papillary Renal Carcinoma (HPRC) h Clinical Features h Bilateral, multifocal papillary renal cell carcinoma h Type I papillary renal carcinoma h Clinical Evaluation – C-Met gene germline mutation testing – Abdominal CT and ultrasound 10
  • 17. B. Met is the Hereditary Papillary Renal Carcinoma gene The gene for type 1 papillary renal carcinoma associated with Hereditary Papillary Renal Carcinoma is the Met proto-oncogene on chromosome 7.14,15 Met is a normal gene (a proto- oncogene) that codes for Met, the cell surface receptor for the ligand hepatocyte growth factor. Met becomes an oncogene when activating mutations occur in the tyrosine kinase domain of this gene in the germline of HPRC patients. A simple blood test is available to determine if the at- risk individual carries the germline Met mutation. Figure 3 Met, the cell surface receptor for the ligand, hepatocyte growth factor, is the gene for Hereditary Papillary Renal Carcinoma. Germline mutations of Met are found in affected individuals in HPRC kindreds. From Linehan, et al 5 11
  • 18. 3. Birt Hogg Dubé (BHD): Chromophobe Kidney Cancer Birt Hogg Dubé is a hereditary syndrome in which affected individuals are at risk for the development of kidney cancer, cutaneous lesions and pulmonary cysts. Figure 4 Birt Hogg Dubé (BHD) is a hereditary cancer syndrome in which patients are at-risk for the development of cutaneous lesions (fibrofolliculoma), pulmonary cysts and kidney cancer. From Linehan et al.5 The kidney tumors can be chromophobe, hybrid/oncocytic, papillary or clear cell RCC.16 The BHD gene was recently identified on chromosome 1717 and a blood test is now available for determination of whether at-risk individuals are affected with BHD.18 Table 5 Clinical Features of Birt Hogg Dubé (BHD) h Cutaneous nodules (hair follicle tumors, fibrofolliculoma) on the face and neck h Pulmonary cysts h Renal tumors h Chromophobe RCC h Oncocytic hybrid RCC h Clear cell RCC h Oncocytoma 12
  • 19. Figure 5 BHD associated kidney cancer can be chromophobe, hybrid/oncocytic RCC or oncocytoma. From Linehan, et al.5 Table 6 Clinical Evaluation-BHD • BHD gene germline mutation testing • Dermatologic evaluation-skin biopsy • Lung CT • Abdominal CT/ultrasound 13
  • 20. 4. Hereditary Leiomyomatosis Renal Cell Carcinoma (HLRCC) A. HLRCC: Clinical Features Hereditary leiomyomatosis renal cell carcinoma (HLRCC) is a hereditary cancer syndrome in which affected individuals are at risk for the development of cutaneous and uterine leiomyomas (fibroids) and an aggressive form of type 2 papillary kidney cancer.19 Figure 6 HLRCC is a hereditary cancer syndrome in which affected individuals are at-risk for the development of cutaneous and uterine leiomyomas and type 2 papillary renal carcinoma. From Linehan, et al.5 Table 7 Clinical Features of Hereditary Leiomyomatosis Renal Cell Carcinoma (HLRCC) h Cutaneous nodules (leiomyomas) h Uterine leiomyoma (fibroids) h Uterine leiomyosarcoma h Renal tumor (type 2 papillary RCC) h Often solitary h Aggressive, early to metastasize B. Fumarate Hydratase (FH) is the gene for HLRCC The Krebs cycle enzyme, fumarate hydratase (FH) is the gene for the hereditary form of Type II papillary renal carcinoma associated with Hereditary Leiomyomatosis Renal Carcinoma (HLRCC) and there is a blood test available to determine whether or not a patient is affected with HLRCC.20 These tumors can be very aggressive21 and partial nephrectomy is often not recommended for these patients. Early intervention with nephrectomy is recommended as these aggressive tumors may spread early. 14
  • 21. Table 8 Clinical Evaluation-HLRCC • Fumarate hydratase (FH) gene germline mutation testing • Dermatologic evaluation-skin biopsy • Abdominal CT/ultrasound • Pelvic CT/uterine ultrasound 5. Familial Renal Carcinoma (FRC) Recently scientists in Iceland have performed studies suggesting that genetic susceptibility may be a major component in the development of ordinary, “sporadic” renal carcinoma. In their initial study, 68% of individuals in Iceland who had kidney cancer had up to a second degree relative (a second cousin) with kidney cancer. This work suggests that it is important to ask all patients with renal carcinoma whether any other family member also was affected with renal carcinoma. Urologic surgeons at the NCI are currently studying families in which multiple members are affected with kidney cancer (FRC) in order to identify the genetic basis of this form of kidney cancer.22 Figure 7 Studies have shown that there may be a strong hereditary component to “sporadic” kidney cancers. Investigators are currently intensively studying families in which more than one family member has kidney cancer in order to determine the gene(s) for familial renal carcinoma. From Linehan, et al.5 15
  • 22. III. Techniques and Indications (Peter Pinto, M.D.) A) Laparoscopic Nephrectomy Surgical Technique: Transperitoneal Approach Positioning The following will describe the operative technique for a transperitoneal left laparoscopic nephrectomy. The patient undergoes general endotracheal anesthesia, prophylactic antibiotics and placement of an orogastric tube and urinary catheter. While supine the lower midline incision is marked with an indelible pen. This is done prior to positioning to allow the incision for possible specimen extraction to be symmetric in reference to the midline. This is difficult to judge after the patient is positioned. The patient is then placed in a modified left flank position with a bump under their left side. The right arm is tucked and the left arm is folded over cushions across the chest. (Picture 1) Picture 1) Positioning for transperitoneal left laparoscopic renal surgery. From Pinto, et al.23 All pressure points are checked and padded. The patient is secured to the table with 3 inch wide silk tape. In order to confirm that the patient is adequately secured, the table is maximally tilted prior to draping. Port Placement The operating table is returned to a neutral position and the patient is prepped and draped in the standard fashion. The surgeon and assistant stand on the patient’s right side and the scrub nurse on the left side by the legs. In cases where a robotic camera holder is used, it is secured to the table at the level of the patient’s right shoulder prior to draping. With the aid of a Veress needle, pneumoperitoneum is established at a pressure of 20 mmHg. Four trocars are subsequently placed: a 10mm periumbilical port for the camera, a 12mm port in the left midaxillary line at the level of the umbilicus, a 12mm port in the midline of the premarked pfannensteil incision, and a 5mm trocar in the midline, halfway between the umbilicus and 16
  • 23. xyphoid. (Picture 2) In obese patients, the port placement is altered slightly. In these cases, the ports are shifted laterally, with the camera port at the level of the umbilicus, but lateral to the rectus muscle. (Figure 1) Picture 2) Port placement for left laparoscopic nephrectomy/partial nephrectomy. Lower midline port can be extended for specimen retrieval. Figure 1) Port placement shifted laterally for obese patient. From Williams et al.24 A visualizing trocar is used to place the first port, allowing the remaining three to be placed under vision. Even in the setting of previous abdominal surgery, initial entry with a veress and visualizing trocar has been shown to be safe. ( J Urol 170, 61–63, July 2003) All trocars are secured to the skin with suture to avoid inadvertent removal during surgery. 17
  • 24. Procedure The table is then tilted maximally to the right to help the bowel fall medially. This can be facilitated by an assistant using a paddle retractor through the low midline port. The retroperitoneal space is entered by incising the line of Toldt sharply from the spleen to the iliac vessels. (Figure 2) Figure 2) Entering the retroperitoneal space As the descending colon is reflected medially care must be taken to develop the plane between the mesocolon and anterior gerota’s fascia. If not, a common mistake is to dissect lateral to the kidney. This divides the renal attachments laterally and makes future hilar dissection more difficult. In order to obtain proper exposure to the renal hilum and especially for upper pole tumors, the splenophrenic ligament must be incised. Take care to avoid inadvertent injury to the stomach and diaphragm as the spleen is being mobilized. Once completely mobilized, the spleen, tail of pancreas and splenic flexure will fall medially out of the operative field. Upper pole dissection is then carried out. The lienorenal ligaments are divided sharply in order to avoid inadvertent splenic lacerations. This dissection is carried out toward the renal hilum in order to separate the adrenal gland and expose the adrenal vein as it enter the renal vein. In cases where the tumor location does not necessitate taking the adrenal gland, the renal vein may be transected lateral, preserving the adrenal vein’s entry into the renal vein. Alternatively, the adrenal vein can be clipped and divided. (Figure 3) 18
  • 25. Figure 3) Upper pole mobilization when needed With the upper pole of the kidney mobilized, attention is turned to the lower pole of the kidney, ureter, gonadal and renal hilum. Dividing the colorenal ligaments allows the colon to fall medially and exposes the lower pole, gonadal vessels and ureter. (Figure 4) Figure 4) Lower pole, ureteral and gonadal dissection A plane is developed between the psoas muscle and the gonadal/ureteral packet. If the ureter is difficult to identify, or obscured by a large lower pole mass, the dissection can be started distally at the level of the iliac vessels and continued proximally to the renal vein. At the level of the renal hilum lumbar vessels are ligated and divided as necessary. The neuro-lymphatic and fibrofatty tissue surrounding the renal artery and vein is ligated and divided isolating the artery down to its take off from the aorta. This dissection also allows for a hilar lymphadenectomy to be performed if indicated. The renal artery and vein can be controlled with titanium or polymer clips (leaving 3 on the patient side) or an endoscopic linear cutter stapler with a vascular staple load (Figure 5). 19
  • 26. Figure 5) Ligation of the renal vessels With the vessels divided, the ureter is ligated and the remaining lateral, posterior and superior attachments are freed with cauterized scissors. If intact specimen extraction is to be performed, the lower midline trocar is then exchanged for an entrapment sac device (15mm Endo Catch II, US Surgical, Norwalk, CT). The specimen can then be placed into the deployed entrapment sac and delivered through a low midline incision which incorporates the lower midline trocar. The extraction incision is then closed and pneumoperitoneum is re-established to ensure that adequate hemostasis was obtained. This is done at a pressure of 5mmHg to reveal bleeding that might be present at normal insufflation pressures. Alternatively, the specimen can be morcellated. In these cases the specimen must be placed in an impermeable entrapment sac (Lapsac® Cook, Bloomington, IN) prior to morcellation. The 12mm trocar sites are then closed with the aid of a needle-point suture passer after the pneumoperitoneum has been evacuated. Indications for laparoscopic radical nephrectomy include organ confined tumors (stage Tl or T2) not amenable to nephron-sparing surgery. Some investigators have demonstrated that even large masses (12cm) can be removed laparoscopically. Tumors spreading beyond Gerota's fascia or involving the renal vein are technically challenging, but have been performed. Laparoscopy has also provided a benefit to patients in the setting of metastatic disease. Surgical Technique: Retroperitoneal Approach Positioning The patient is placed on the operating table in the standard flank position with the side of pathology facing up. The top arm is draped over a padded Mayo stand or multiple pillows. An axillary roll is placed. The bottom leg is flexed and bent while the top leg is straight. The kidney rest is elevated and the table is flexed in order to maximize the working space between the ribs and iliac crest (Picture 3). 20
  • 27. Picture 3) The patient is placed in a full flank position with the kidney rest elevated and the table flexed. This increases the distance between the costal margin and iliac crest, thus maximizing the working space. The skin between the iliac crest and the ribs should be taut on palpation. All pressure points are padded and the patient is then secured to the table with tape at the shoulders, hips, and legs. The table is rotated left and right to make sure the patient does not shift during the procedure. The surgeon and camera assistant are positioned facing the patient's back. The scrub nurse stands on the opposite side at the end of the table. (Fig 6) Fig 6) Operating room setup for laparoscopic retroperitoneal renal surgery. From Pinto et al.25 21
  • 28. Two monitors are used to provide all personnel with an unobstructed view. Alternatively, the camera assistant can be replaced by a mechanical or robotic arm which can be fixed to the operating table to hold the camera. The robotic arm is controlled by the surgeon through voice commands, a hand control or a foot pedal. Access and Port Placement Three trocars are positioned in the anterior, mid, and posterior axillary lines.(Picture 4) Picture 4) Trocar placement. The laparoscope is positioned in the mid-axillary line. A 12-mm port is placed in the posterior axillary line. A 5-mm port is placed in the anterior axillary line. Initial entry into the retroperitoneal space is established off the tip of the 12th rib via the open Hassan technique. A 1.5 cm transverse incision is made just anterior and inferior to the tip of the 12th rib. The incision is carried down sharply through the posterior thoracolumbar fascia, flank muscles and anterior thoracolumbar fascia. Upon entering the retroperitoneal space, the index finger is used to sweep the peritoneum anteriorly and medially, developing a space between the psoas muscle anteriorly and Gerota’s fascia posteriorly. (Fig 7) 22
  • 29. Fig 7) The surgeon's finger is used to start the retroperitoneal dissection. A space behind the kidney is created to place the balloon dissector. See Pinto et al.25 The retroperitoneal space can then be created with balloon dissection (Fig 8A) and the balloon tipped 10mm trocar (Fig 8B) provides a seal to establish the pneumoretropertioneum. A) B) Fig 8 A) The working space is created by balloon dissection. The kidney and peritoneum are displaced anteriorly exposing the renal hilum. B) Blunt-tipped trocar. The collar on the port slides down against the balloon tip to prevent loss of pneumoretroperitoneum. See Pinto et al.25 Alternatively, retroperitoneal access can be obtained directly with a 12 mm laparoscopic visualizing trocar. Once the characteristic fat of the retroperitoneum is seen, insufflation is begun. The laparoscope is used to bluntly develop the working space. Surgical Technique Renal dissection is carried out as was described previously, but without the need to manipulate the peritoneal organs. The psoas muscle and ureter are the initial structures for orientation, since the intraperitoneal structures are not seen. Maintaining orientation is important throughout the procedure. This helps identify landmarks such as the psoas muscle, peritoneal 23
  • 30. reflection, ureter, gonadal vein, and pulsations of the renal artery. Initial dissection is carried out posteriorly to isolate the ureter and mobilize the lower pole. The peritoneal attachments anterior to the kidney are not divided in order to prevent the kidney from falling into the working space. Blunt dissection along the ureter reveals the gonadal vein. Theses structures are traced cephalad to the renal hilum. Dissection along the aorta or vena cava will quickly bring the renal vessels into view. The renal artery is encountered first as it lies posterior to the vein. Hilar ligation is performed as was described in the transperitoneal approach. Once the vessels have been controlled, the anterior surface of the kidney is mobilized from its peritoneal attachments. Care is taken not to enter the peritoneum and injure the adjacent bowel and mesentery. Post-Operative Management Prior to extubation the orogastric tube is removed. After routine observation in a post anesthesia care unit, the patients are transferred to a non-monitored floor bed. In the morning of the first post operative day, the urinary catheter is removed, routine blood work is performed, ambulation is encouraged and oral liquids are started. The diet is then advanced as tolerated throughout the day. Routine discharge is in the morning of post-operative day # 2 if blood work and vital signs are stable, the patient is able to ambulate and tolerate oral diet. 24
  • 31. III. Techniques and Indications: B) Laparoscopic Partial Nephrectomy Surgical Technique: Transperitoneal / Retroperitoneal Approach Positioning and Port Placement Same as for laparoscopic nephrectomy. Additional instruments needed for laparoscopic partial nephrectomy include: laparoscopic vascular clamps (Picture 5- bulldogs, Picture 6- Statinksy), ultrasound with a laparoscopic transducer and topical hemostatic sealant. Picture 5) Laparoscopic bulldog clamps Picture 6) Laparoscopic Statinsky clamp Procedure When the renal mass is abutting the collecting system, and entry into a calyx is expected, cystoscopy and placement of an open ended 5 French ureteral catheter is performed before positioning. This aids in reconstructing the collecting system by retrograde injection of dilute methylene blue. As in open surgery, dissection of the kidney is performed leaving a “cap”of fat overlying the renal tumor. Intraoperative ultrasound aids in determining the tumor’s depth and also to rule out any additional tumors. 25
  • 32. Exophytic masses that are attached by a narrow base and peripherally located can be excised without the need for vascular occlusion. The capsule around the tumor is scored with cautery, and the mass is removed with the aid of laparoscopic scissors or an ultrasonic scalpel. Hemostasis is obtained with the aid of an argon beam coagulator and hemostatic sealants. Large, more broad-based or deeper tumors require interruption of renal blood flow. Prior to clamping the renal hilum, a brisk diuresis is initiated with the use intravenous fluids and osmotic diuretics. Mannitol (25 grams) is given when the surgeon begins to free up the renal hilum allowing for increased perfusion of the renal parenchyma along with protection from free radical deposition. The capsule around the mass is scored, and a laparoscopic bulldog clamp is placed on the renal artery. Alternatively, a laparoscopic Statinsky vascular clamp can be used, but this requires the placement of an additional trocar. Hilar clamping is performed to allow for a bloodless field and therefore better visualization when resecting the tumor. The tumor is excised with cold scissors to allow pathological assessment of the tumor’s margins. The tumor is excised ensuring a rim of normal parenchyma around it. Any suspicious areas in the renal bed can be biopsied and sent for frozen section analysis. Retrograde instillation of methylene blue via the ureteral catheter ensues that the collecting system has not been entered. If it has, repair is performed with 2-O polyglactin suture. Open vessels seen in the renal bed are suture ligated with 2-O polyglactin suture. A gelatin matrix hemostatic sealant is applied and the kidney is reconstructed by reapproximating the renal defect over surgicel bolsters (Picture 7) using O polyglactin sutures through the renal capsule. Picture 7) Surgicel pledgets, approximately 1 cm wide and 4 cm long, are prepared prior to tumor excision. The clamps on the renal vessels are removed and the kidney is observed to confirm it is being well perfused and there is no active bleeding. Prior to exiting the abdomen, a closed suction drain is left in the retroperitoneum behind the kidney. Cold ischemia is technically feasible laparoscopically, but not practical. Therefore when operating under the time constraints of warm ischemia, it is important to be expeditious in your approach to resection and reconstruction of the kidney. Indications for laparoscopic partial nephrectomy are as described for the open procedure. Although initially indicated for tumors involving a solitary kidney or bilateral disease, nephron- sparing surgery is now performed for tumors smaller than 4cm in the presence of a normal contralateral kidney. 26
  • 33. IV. Techniques and Indications: Robotic Assisted Nephrectomy and Partial Nephrectomy Nephron sparing surgery for complex renal tumors, such as multiple, endophytic, or hilar tumors, may further preclude a laparoscopic approach for most urologists and may pose challenges for even experienced laparoscopic surgeons. A renal mass with tumor thrombus may require vascular suturing, which is also challenging for laparoscopic techniques. It is in these cases that robotic renal surgery may be of assistance. Potential advantages include 3-dimensional stereoscopic vision, articulating instruments, and scaled-down movements reducing tremor. The articulating instruments and increased freedom of movement may also allow the surgeon to replicate well established open surgical maneuvers more readily. This technique emulates both open and laparoscopic techniques of renal cancer surgery. (Rogers et al. 2008) Surgical Technique: Transperitoneal Approach Positioning, Ports, and Docking: After intubation, orogastric tube placement and bladder catheterization, the patient is positioned in modified lateral position. Full flank position may also be used. Following abdominal insufflation with a Veress needle. Ports are placed as demonstrated in Picture 8. A 12mm periumbilical port is placed for the camera. Two robotic instrument ports are placed approximately 8 cm from the camera in a wide “V” configuration centered on the renal tumor. These ports may be shifted laterally and/or superiorly for patients with a large body habitus or upper pole tumor location. A 12 mm assistant port is placed inferior to the camera port. An optional 5mm assistant port may be placed above the camera port if needed. For docking (as seen in Picture 9), the robot is brought in posteriorly at approximately a 20 degree angle toward the head of the patient. Picture 8) Port placement for left sided robotic renal surgery 27
  • 34. Picture 9) daVinci robot is docked against the patient’s back at approximately a 20 degree angle toward the head of the patient. Bowel Mobilization: A zero degree lens is used initially, but a 30 degree downward lens may also be utilized as needed. Robotic instruments used include a Bipolar Maryland forceps, monopolar cautery scissors, and needle drivers. The peritoneum is incised sharply along the Line of Toldt and the bowel is mobilized medially using sharp and blunt dissection, developing the plane between the anterior Gerota’s fascia and the posterior mesocolon. The bedside assistant maintains medial counter-traction. Dissection is continued along the upper pole of the kidney to mobilize the spleen or liver. We utilize robotic assistance from the beginning of bowel mobilization unlike some reports of a “hybrid technique”, initially utilizing conventional laparoscopy to reflect the bowel. Anatomical Landmarks and Hilar Dissection: Continued medial reflection of the bowel allows for exposure of the gonadal vessels and the ureter. These structures are retracted anteriorly, exposing the underlying psoas muscle. Care is taken not to strip the fascia from the psoas muscle. Dissection then proceeds towards the renal hilum. The Maryland bipolar forceps are used to place the kidney on stretch and the renal hilar vessels are dissected to allow access for clamp placement. Lateral renal attachments are left in place to aid in countertraction. Venous branches can be ligated as needed for exposure Ultrasound and Tumor Exposure: A laparoscopic ultrasound probe is used to map the location and size of renal tumor(s) and to confirm resection margins and depth during partial nephrectomy. Gerota’s fascia is opened and the fat is cleaned off the renal capsule to expose the tumor(s). The use of color Doppler may be used to identify adjacent vessels. The margin of resection is scored 28
  • 35. circumferentially using monopolar cautery. In cases of nephrectomy and tumor thrombectomy, it is used to determine the extent of tumor thrombus. Hilar Clamping, Tumor Excision and Renal Reconstruction during Partial Nephrectomy: For tumors that are endophytic or adjacent to the renal hilum, resection is done under warm ischemia. The assistant clamps the renal hilar vessel(s) using laparoscopic bulldog clamp(s) through the primary 12mm assistant port. Mannitol (12.5 gm) may be administered intravenously prior to clamping. The tumor is resected along the previously scored margin using cold resection with the robotic monopolar scissors. The Maryland bipolar forceps are used to manipulate the tumor for exposure and to aid in dissection. The assistant uses suction to expose and maintain visualization of the resection plane of the tumor. After excision, the tumor can be placed beside the kidney or on top of the liver for later retrieval. If the status of the surgical margin is in question, a biopsy and frozen section analysis may be performed. Hemostasis is achieved using a combination of cautery, hemostatic agents, and suturing. A pre-placed ureteral catheter may be used to inject methylene blue to identify entry into the collecting system. The robotic instruments are exchanged for needle drivers. A 3-0 Vicryl suture on an RB-1 needle is used to achieve hemostasis and repair any previously identified entry into the collecting system (a SH needle may also be used). Sutures may be secured with either absorbable suture clips or by tying knots. Renal parenchymal defects are approximated over surgicel bolsters using 2-0 Vicryl sutures on an SH needle (a 0-Vicry suture on a CT-1 needle may also be used). A hemostatic agent is applied. Preplacing surgicel bolsters and sutures in the abdomen may reduce warm ischemia time during earlier experience. The kidney is placed back on stretch using the robotic needle driver and the hilar clamp is removed by the assistant. Hemostasis is confirmed. A pre-placed lap pad may be used to apply pressure to the resection site. The specimen is placed in a retrieval bag and removed through the primary assistant 12 mm port, enlarging the port site if needed. Gerota’s fascia is approximated over the defect using a running 3-0 Vicryl suture on an SH needle. A drain is placed in the perinephric space. 26-54 29
  • 36. V. Role of Targeted Systemic Therapy in Management of Advanced Clear Cell and non-Clear Cell Kidney Cancer (Jeffrey A. Sosman, M.D.) Introduction Therapy of advanced Renal Cell Carcinoma (RCC) has gone through a major evolution over the past few years. Previously, patients with RCC were considered for high dose Interleukin-2 therapy, if young and healthy and IFN-alpha for the less robust or elderly patient. A relatively small subset of patients did benefit from cytokine therapy, but far too few to effect median progression-free (PFS) or overall survival (OS)55,56. The great majority of patients only suffered the toxicity of treatment. This all began to change following the remarkable work of Drs. Kaelin, Maxwell, Linehan and others who helped define the role of the VHL gene and the consequence of loss of its function.57 The generation of an antibody to the vascular endothelial growth factor, VEGF (anti-VEGF; Bevacizumab) molecule provided an initial tool to interrupt the downstream effects of VHL (Von Hippel Lindau) loss. The stabilization of the HIF-1a and HIF-2a molecules with the subsequent over expression of VEGF, PDGF, EGFR, CAIX, EPO, Glut1 etc was key to the malignant phenotype of clear cell RCC. This is a critical pathway not simply in genetic syndromes with Von Hippel Lindau, but in at least the majority of patients with sporadic clear cell RCC. In the initial report by Yang et al., anti-VEGF was shown to increase PFS >2 fold in a cytokine (IL-2) refractory population even though its objective response rate was only 10%.58 However, it was clear that even those without objective clinical responses had a far different clinical course than those on placebo with many having prolonged stable disease or small degrees of regression not meeting the 30% RECIST threshold of an objective response. A reassessment of meaningful clinical responses needed to be performed the first time there may have been convincing data that stable disease was an indication of benefit in RCC. In the interim, a number of multi- targeted tyrosine kinase inhibitors (TKI) that targeted the VEGFR2 tyrosine kinase as well as a variety of other receptor signaling pathways, have been evaluated and some approved for therapy of advanced RCC. These orally administered drugs were also able to inhibit other receptors including VEGFR1, VEGFR3, PDGF, c-kit, and RAF to name a few. The initial two agents receiving the most attention and most rapidly approved by the FDA were BAY43-9006 (Sorafenib) Nexavar and SU011248 (Sunitinib) Sutent.59 While their spectrum of kinase targets was overlapping, there were some key differences. Sorafenib inhibited RAF, while Sunitinib was a more potent and effective inhibitor of VEGFR2. (See Table 1) 30
  • 37. Table 1: Phase II/III Trials of Targeted Agents in Untreated RCC patients Phase Pt Response Median Median Drug of Risk factors Nephrectomy Number Rate PFS OS Study 375 Favorable(34%) 39% 11 mos NA Sunitinib vs IFN III 90% 375 Intermediate(56%) 8% 5.1 mos NA Bevacizumab + 327 Favorable (28%) 31% 10.2mos NA III 100% IFN vs IFN 322 Intermediate(56%) 12% 5.4 mos NA Temsirolimus 209 8.6% 5.5mos 10.9mo Poor Prognosis Temsirolimus+IFN III 210 67% 8.1% 4.7mos 8.4 mo (74% MSKCC) Vs IFN 207 4.8% 3.1mos 7.3 mo Sorafenib 97 Favorable NA(68%)* 5.7mos NA II 95%/98% Vs IFN 92 Intermediate NA(39%)* 5.6mos NA Number in parenthesis represents % of patients with some degree of tumor shrinkage, not an objective response rate A) Sorafenib Initially, Sorafenib demonstrated prolonged progression-free survival in cytokine refractory patients in a phase II randomized trial where stable disease patients at 12 weeks were randomized to continue Sorafenib or switched to placebo (randomized discontinuation).59 Those patients having switched to placebo had a significantly less PFS than those on Sorafenib, when the placebo patients who progressed were rechallenged with Sorafenib, stable disease could be re-established. This was confirmed and proven in a large phase III trial of Sorafenib versus placebo in cytokine-refractory patients where again as in the phase II trial there was a doubling in progression-free survival from 2.8 to 5.5 months9. Unfortunately this study was then unblinded (per recommendations of the FDA) and overall survival results were confounded by the significant cross-over of patients from placebo to Sorafenib. It is worth pointing out that in both of these studies the objective response rate of Sorafenib was < 10% while many patients had smaller degrees of regression and prolonged stable disease. Later after its approval a relatively small, 189 patient, randomized phase II trial of Sorafenib or IFN in therapy-naïve patients was conducted. Surprisingly, there was no advantage to the Sorafenib arm.60 This was unexpected based on the second line results with Sorafenib in cytokine-refractory patients. While a low objective response rate could have been expected, the lack of any improvement in patients’ PFS with a median of 5.7 months, was a surprise. The trial also examined the effect of cross over to Sorafenib and dose escalation of Sorafenib in patients who progressed on their initial therapy. Patients progressing on IFN, could receive Sorafenib at standard doses of 400mg BID. This group of patients did exhibit some stabilization of their disease for 5.7 months and some degree of tumor shrinkage in 75% of patients. Those who progressed on Sorafenib could be dose escalated to 600mg BID and among these 44 patients, a 4.1 months median PFS and tumor shrinkage in 44% were observed. For all practical purposes, this large randomized phase II trial signaled the end of Sorafenib as a front line therapy in the 31
  • 38. majority of patients. Larger, single agent, upfront trials with Sorafenib at standard doses, are unlikely to be explored further. B) Sunitinib The pathway to Sunitinib approval was quite different. Initial phase II studies did demonstrate a high response rate of 35-45% in cytokine-refractory patients, as well as prolonged progression-free survival. Following this result, Sunitinib was directly compared to IFN-alpha in therapy naïve patients. This large 750 patient trial demonstrated a remarkable objective response rate for Sunitinib that remained very high at 40%, while IFN's response rate was <10%8. Furthermore, the median progression-free survival was 11 months, more than double that of IFNα. The difference in overall survival did not meet significance due to the lack of events at the time of the analysis. Again the survival analysis is confounded by patient crossover from IFN to Sunitinib as allowed in the protocol once Sunitinib was approved by the FDA. C) Bevacizumab Following the approval of both MTKI, the results of several phase III trials featuring Bevacizumab (anti-VEGF) have been reported. In a large study now published in Lancet by Escudier, et al., IFN alone was compared to IFN with anti-VEGF in a population of therapy- naïve RCC patients.61 The results showed a striking improvement in response rate (31% vs 7%) and progression-free survival (10.2 mos vs 5.4 mos). The response rate was better than seen in both the initial Yang trial in cytokine refractory patients (10%) and the 13% in the randomized phase II trial of anti-VEGF +/- erlotinib (EGFR TKI). It is possible that the addition of IFNα to anti-VEGF led to a better response rate and improved the progression-free survival of anti- VEGF. But there are no more definitive results to either support or counter this argument. Finally, limited release results from a trial conducted by CALGB show there were further supporting data to the benefit of Bevacizumab plus IFN. The trial represented another phase III trial of IFN +/- anti-VEGF. The combined arm (anti-VEGF + IFN) demonstrated an improved response rate and progression-free survival compared to IFN alone. Final results are still pending. Issues with phase III trials in front line therapy When reviewing the important trials with Sorafenib, Sunitinib, and Bevacizumab, there are several important issues needing to be addressed. First, patients on all of these studies were either intermediate or low-risk based on the MSKCC criteria62. They had 0-2 of (anemia, hypercalcemia, high LDH, lower performance status (<70% Karnofsky criteria) or either within 1 year of diagnosis of RCC or without nephrectomy) the five criteria. Three or more of these criteria were indicative of poor prognosis. In previously evaluated patients at MSKCC, this criteria could separate patients with a 20 month median (low risk, favorable prognosis) overall survival to as low as a 4 month median survival for those high risk (poor prognosis) patients. The entry criteria for all of the above studies also required clear cell histology. All patients on the Bevacizumab plus IFN trial had nephrectomy (as per entry criteria), while over 90% had prior resection of their primary on the Sunitinib trial. There were a small number of poor prognosis patients entered on both trials, 6% on the Sunitinib trial and 8% on the Bevacizumab + IFN trial. Both the front and second line trials with Sorafenib enrolled patients with intermediate to low risk and nearly 90% had nephrectomy. The results in the poor 32
  • 39. prognostic patients were dismal but PFS may have been improved from 1.2 months to 3.7 months in those receiving Sunitinib. Not even this small effect was seen in the Bevacizumab+ IFN trial where poor prognosis patients had a 2.1-2.2 median progression free survival regardless which arm they were enrolled on. D) Temsirolimus In phase I and II clinical trials, RCC patients appeared to derive a benefit of CCI-779, Temsirolimus, even though the response rates were less than 10%. Patients improved with less bone pain, improved appetite and weight gain. Temsirolimus is a mTOR (mammalian target of rapamycin) inhibitor. mTOR is downstream from PI-3/Kinase/ Akt. It represents a pathway that can modulate protein synthesis (translation) including that of HIF-1a and HIF-2a. The phase II trial demonstrated that patients with poor prognostic factors had the greatest benefit. Benefit was based on a major extension in progression-free and overall survival in the poor prognosis group.63 Overall survival was much better than expected for this poor prognosis patient group. This led to a phase III trial comparing Temsirolimus versus IFN in poor prognostic patients (3 or more of 5 criteria in MSKCC). The trial did get slightly modified to increase poor accrual by adding one more poor prognostic factor, multiple disease sites (by organ involvement). Patients needed 3 or more of the 6 risk factors for entry. There were no restrictions on histology and requirements for nephrectomy. About 20% had non-clear cell histology and about 33% had their primary in place and were enrolled within their first year of diagnosis. The trial was an ambitious effort in a poor prognostic group of patients who in the past would likely not have been treated with cytokines due to their overall performance status. The trial included 3 arms; one with IFN alone; one with Temsirolimus; and the other a combination of both IFNa plus Temsirolimus 64. The doses of Temsirolimus were attenuated to allow concomitant IFNa therapy. The trial’s primary endpoint was overall survival, with progression-free survival only a secondary endpoint. Overall survival was increased from a median of 7.3 to 10.9 months with an overall hazard ratio of .73 and the p value=0.0069. PFS was improved in both the Temsirolimus arm and the IFN plus Temsirolimus arm, but those in the combined arm did not get any improvement in overall survival. This may have to do to the extensive dose modification of when IFN and Temsirolimus were given together. This result clearly established Temsirolimus as the standard therapy for poor prognostic patients. Overall response rates in all three arms were under 10%, further evidence that objective responses were a poor indicator of treatment benefit from drugs in RCC. Several additional analyses were done retrospectively on pre-treatment patient characteristics and clinical outcome. For those 74% of the overall enrollment who did qualify under the MSKCC model as a poor prognostic group (3 or more of 5 MSKCC criteria) the benefit from Temsirolimus was most prominent. Most of the benefit was apparently concentrated within this group of patients. Furthermore, about 20% of patients had histology of their tumor different from the typical clear cell. Papillary renal cancer makes up the bulk of these non-clear cell cancers. Others include chromophobe and sarcomatoid (high grade IV) that make up a small number of cases. In those patients with non-clear cell histology the benefit was especially prominent as is described below in non-clear cell RCC. This was unique to this trial of Temsirolimus since the other upfront trials excluded these patients and were restricted to clear cell RCC alone. 33
  • 40. Toxicities from the targeted agents (See Table 2) In addition to overall survival, progression-free survival, and objective response rate, toxicity, and patient tolerance is of critical importance when considering treatment with this class of agents. This is especially true, since the nature of these agents is to continue their administration indefinitely or until progression. Complete responses are very rare and much of therapy is administered with the intent to maintain disease control. Both Sunitinib and Sorafenib have the most overlapping spectrum of toxicities as might be expected based on the targets of their action. Table 2: Toxicities from Targeted Agents Toxicities HTN Proteinuria Vascular Stomatitis HFS Rash Diarrhea Fatigue Complications Sunitinib X X X x X X Sorafenib X x X X X X Temsirolimus x X X Bevacizumab X X X X Common and significant problem x Less common and/or infrequent problem Sunitinib toxicity Sunitinib can cause fatigue, hypertension, a functional (symptomatic with minimal findings on examination) stomatitis, diarrhea, and some hand-foot syndrome. Most common grade 3 or 4 toxicities have been due to fatigue and hypertension, though hypertension in general can be well controlled with medications. At times multiple anti-hypertensive agents are required for adequate blood pressure control. Hypothyroidism is becoming more appreciated as a frequently associated side effect. Thyroid functions should be followed throughout treatment. Sometimes the fatigue from Sunitinib is due to thyroid dysfunction and this can be easily addressed with hormone replacement. A less frequent but increasingly recognized toxicity is a decline in cardiac ejection-fraction. While declines below normal range or greater than 10% of baseline are relatively common at 10-15%, symptoms of congestive heart failure is seen in less than 5% of patients. Obtaining baseline ejection fraction and subsequent follow-up cardiac evaluations are now recommended even in the absence of symptoms. Sorafenib toxicity Sorafenib toxicities include many of those seen with Sunitinib but there is a slight but recognizable change in the spectrum of them. Hypertension is common as with Sunitinib, but fatigue is infrequently a dose limiting toxicity. Diarrhea is experienced in up to 40% of patients but is grade 3 or 4 in only several percent. Most bothersome has been the hand foot syndrome that is observed in up to 30% of patients. While severe desquamation of the hands and feet is uncommon, symptomatic callus formation with neuropathic pain is frequently observed. Patients can have trouble with walking and use of their hands. This toxicity is readily reversible upon suspending drug administration, but if the symptoms worsen while drug continues to be given then recovery can be quite slow. Sorafenib can also induce a number of different rashes from a diffuse macular, papular, pruritic rash to a localized acneform rash similar to that associated with EGFR tyrosine kinase inhibitors or antibodies. It can also cause multiple furuncles and 34
  • 41. carbuncles in and around the axillas or the inguinal regions. These can become secondarily infected and required drainage and antibiotics. Bevacizumab toxicity Bevacizumab is very well tolerated on a daily basis. Minor fatigue and myalgias can be associated with intravenous administration for 24-48 hours afterwards. Patients frequently experience hypertension and proteinuria. Blood pressure control is important since lack of control can lead to hypertensive crisis. Proteinuria must be monitored closely and in general is very reversible. Recommendations have become more lax in terms of the threshold for proteinuria in deciding to hold Bevacizumab. At the present time, 3.5 gm of protein in a 24 hour urine is the limit for continuing Bevacizumab dosing. Otherwise the dose is held until protein in urine has dropped below the threshold. Bevacizumab is generally safe and well tolerated. It must be recognized that Bevacizumab can significantly slow wound healing and even lead to wound dehiscence. This must be considered around procedure and any major surgery. In general Bevacizumab should be held two weeks prior to surgery and at least 4 weeks following surgery to allow healing to take place optimally. However there are much more infrequent toxicities of a very serious nature. These include vascular events, especially arterial thrombotic and venous thromboembolic , hemorrhage, multifocal leukoencephalopathy associated with poorly controlled hypertension, gastrointestinal perforation, and fistula formation. Temsirolimus toxicity Finally, Temsirolimus is tolerated quite well. Some degree of asthenia (fatigue) is relatively common. However, this can be difficult to attribute to either disease or therapy. Additionally, patients have some degree of pancytopenia with anemia being most prominent. Secondary infection or bleeding is very rare. Some mouth ulcerations and rashes can also be seen. An untoward toxicity relatively unique to this agent is hyperglycemia, hypertriglyceremia, and hyperlipidemia. Pancreatitis may occur secondary to the elevated tryglycerides. While therapy can be cumbersome with its weekly intravenous schedule, its overall toxicity is mild even in the overall more debilitated patients who have been studied. Alternate Doses and Schedules For two of the agents, alternative dosing and scheduling have been attempted. Sunitinib can be given continuously at 37.5mg po daily. This schedule avoids the break in treatment for 2 weeks where conditions may favor regrowth. However, the lower dose, continuous schedule was not superior and the overall response rate appeared lower than the standard 50mg daily for 4 of 6 weeks. This schedule was not directly compared to the standard schedule in a randomized trial. Additionally, toxicity was similar with the continuous dosing schedule. Attempts have been made at dose escalation of Sorafenib. There was some evidence from the randomized phase II trial of Sorafenib that those who progressed on 400mg BID could be dose escalated to 600 mg BID. Amato and his colleague have attempted to follow a very aggressive dose escalation in treatment naïve patients. In 44 patients, 41 were able to be escalated to 600mg BID after 1 month and then 32 of these patients were able to be increased to 800mg BID after 2 months65. There was a very high response rate of 55% with 16% complete responses. Though the median PFS was just 8+ months and overall survival was at 11.5 months. This is surprisingly short considering the high response rate and suggests that these are short clinical responses that may be limited by toxicities. This must be reproduced in a multi-center phase II study, but if positive 35
  • 42. it would be quite significant and ways to ameliorate Sorafenib toxicities would take on even more importance. Until then these results should be looked at with great caution and only considered in a trial setting. Therapy in Patients Who Have Failed Front Line Anti-angiogenesis Therapy There is little information to guide therapy in patients failing their initial treatment. One study has provided some direction in patients who have failed Bevacizumab66. These patients with progressive disease while on Bevacizumab were then treated with Sunitinib. Therapy had to be initiated within several months of stopping Bevaciumab treatment. Of 61 patients there was an objective response rate of 23% with a median progression-free survival of 30.4 weeks (7.4 mos) and overall survival of 47.1 weeks (10.9 months). Of the 57 patients with post-treatment tumor evaluations 51 (84%) demonstrated some degree of tumor regression. Patients who had failed to have any prior response to Bevacizumab were among those who demonstrated objective responses to Sunitinib (11/14 patients with PR). To date, this represents the most mature study of second line therapy. Rini, et al. reported on a cohort of 62 patients who had failed Sorafenib and were subsequently treated with another multi-targeted kinase inhibitor, Axitinib with potent activity against all three VEGFR1, 2,and 3.67 Many of these patients had not only failed Sorafenib, but also Sunitinib, cytokines, and chemotherapy. Of 62 patients, 21% experienced a partial clinical response and 34% had stable disease for at least 3 months. Hypertension as was previously observed in frontline studies was the most problematic side effect from Axitinib. Finally, Escudier has reported on 90 patients from France who had previously received Sorafenib and following progression were switched to Sunitinib (68 patients) and 22 patients who had failed Sunitinib and were then switched to Sorafenib therapy. Among those 68 patients who were switched to Sunitinib there was a 15% objective response rate with an additional 51% with stable disease. The median PFS was 25 weeks. For the 22 patients who had failed Sunitinib and switched to Sorafenib, there was a 9% response rate with stable disease in 55% and median PFS of 17 weeks. While this was only a retrospective study it does support the lack of complete cross resistance for these two drugs even with their similar target profile. Combination Targeted Therapy in Advanced RCC A number of studies have been reported combining several of the active targeted agents including Sorafenib or Sunitinib with Bevacizumab and Temsirolimus with either Bevacizumab, Sorafenib or Sunitinib. While most of these studies enrolled small numbers of patients and included several dose levels, there have been some common findings. In general toxicity of the two agents appears to be exacerbated. There has been an increase in known toxicities such as hand-foot syndrome, hypertension, or proteinuria. In some cases, new toxicities have occurred such as a microangiopathy associated with hemolysis, thrombocytopenia, renal insufficiency and mental status changes. That has been recently observed with Sunitinib and Bevacizumab combination. While some combinations appear to allow full doses of both agents (Bevacizumab and Temsirolimus) most of the combinations required dose reduction of either one or both of the agents. Combinations of Sorafenib and Bevacizumab have required significant dose reductions of both agents. Activity in many cases appears to be preserved but still no formal phase II studies 36
  • 43. have been undertaken with any of the combinations. A recently opened randomized phase II trial will explore three of the combinations of Sorafenib with Bevaciumab, Sorafenib with Temsirolimus, Bevacizumab with Temsirolimus, and as a control, Bevacizumab alone. Additionally, another phase III trial is being planned to compare Bevacizumab with Temsirolimus or with IFN. In general, there remains many questions about combination therapy, especially in the face of toxicity and requirements for lower doses of the agents when they are combined. So far complete responses have been as rare as with single agents. It may be in the future that sequential therapy will be considered the best approach to treatment as it is frequently done today. Only through well performed trials will we gain this information. Use of Targeted agents in the treatment of Papillary Renal Cell Carcinoma (See Table 3) The role of HIF and its hypoxia induced gene expression program in papillary RCC is not understood. In the past most medical urologists, believed these malignancies responded to chemotherapy based regimens. This was never adequately studied to draw any conclusions. Probably the most convincing information about treatment efficacy in papillary RCC comes from the front line Temsirolimus trial as described above. In this trial out of 626 patients about 19% had non-clear cell histology. Of those where histology was better described 75% had papillary RCC as would be expected. In the two arms of Temsirolimus versus IFN there were 73 patients with non-clear cell histology. In a striking result, the progression-free survival was 7.0 versus 1.4 months favoring the Temsirolimus arm and overall survival was more than doubled in those receiving Temsirolimus with OS of 11.6 months versus 4.3 months for IFN–treated patients. This data has a number of problems; the number of patients is small and the actual with papillary RCC is not definite, but assuming the arms are balanced and most are papillary cancers, these results suggest Temsirolimus is an active drug in papillary RCC. Finally, in a retrospective report recently published from 5 centers in the US and France, the clinical outcomes of Sorafenib and Sunitinib in patients with papillary RCC was reported. Forty-one of the 53 patients had papillary RCC and the response rate was only 4.8% (2/41) and both received Sunitinib. The median PFS for papillary RCC treated with Sunitinib was 11.9 months (13 patients) while for the other 28 patients treated with Sorafenib the median PFS was 5.1 months. Few patients had central pathology review. While the data on Sunitinib is limited with only 13 patients, there appears to be some activity (2/13 patients with PR) and PFS was 11.9 months. On the other hand, therapy with Sorafenib had only a median PFS of 5.1 months and no clinical responses in the 28 patients. It is obvious further information is needed to make any definite decisions on therapy with either Sunitinib, Sorafenib or even Temsirolimus. It is critical to perform large enough multicenter studies with only papillary RCC to better gauge effects of any of the above agents. 37
  • 44. Table 3: Experience with Targeted Agents in Papillary Renal Cell Carcinoma Drug Histology Patient Response Median PFS Median OS Number Rate Sorafenib Papillary 28 0/28 5.1 mos NA Sunitinib Papillary 13 2/13 11.9 mos NA Temsirolimus Non-clear cell# 37 NA 7.0 mos 11.6 mos Vs IFN 75% Papillary 36 NA 1.4 mos 4.3 mos # Patients all had poor prognostic factors based on MSKCC and Multiple Organ Sites (>3/6 risk factors) Summary (See Table 4) Great progress has been made in a few years as shown in Table 4. Frontline data supports the use of either Sunitinib alone or Bevacizumab with IFN in patients with favorable or intermediate prognostic factors. The role of nephrectomy is not known, but almost all patients enrolled onto previous studies did have nephrectomies. Those patients with poor prognostic factors should probably receive Temsirolimus. Data on Sunitinib is simply not very convincing at this point but there very well could be activity comparable to Temsirolimus. Second line cytokine failures would be appropriate candidates for Sorafenib based on its prolongation in median PFS. Finally those who have failed front line targeted therapy have very little guidance about what treatment should be given next. Bevacizumab failures can receive a significant benefit from Sunitinib and patients who fail one of the MTKI, Sorafenib or Sunitinib can respond with objective clinical responses or stable disease from the other agent. However, this is an area which still requires extensive study. Lastly, patients who are young, healthy, have good organ function, an excellent performance status, and access to an experienced IL-2 center may be considered for high dose Interleukin-255 . It is the only treatment which offers a chance for cure and very prolonged disease free periods. While this number is less than 10%, because of its impact on these patients is so great, some may consider it for frontline therapy and targeted agents after failing IL-2. We have impacted on many more patients that any of the cytokines ever did. While few studies show an overall benefit except for Temsirolimus, those of us taking care of patients have experienced living for a number of years with disease control where in the past their survival was measure in months to a year. This impact will become more clear with time. Hopefully over that time we will gain further insight how to use these agents. 38
  • 45. Table 4: Potential Standards for Advanced Stage Clear Cell RCC Setting Therapy First-Line Good or intermediate Sunitinib High-dose IL-2 Therapy risk* Bevacizumab? With IFN? Poor risk* Temsirolimus/ Sunitinib Second- Prior cytokine Sorafenib Line Prior VEGFR Temsirolimus?? Therapy inhibitor Prior mTOR inhibitor No data available 39
  • 46. References 1. Jemal, A., Siegel, R., Ward, E., Murray, T., Xu, J., and Thun, M. J.: Cancer statistics, 2007. CA Cancer J Clin, 57: 43, 2007. 2. Linehan, W. M., Bates, S. E., and Yang, J. C.: Cancer of the Kidney. In: Cancer: Principles and Practice of Oncology 7th ed., 7th Edition ed. Edited by DeVita, S. E., Hellman, S., and Rosenberg, S. A.: Philadelphia: Lippincott Williams & Wilkins, chapt. 30, pp 1139-1168, 2005. 3. Linehan, W. M., Vasselli, J., Srinivasan, R., Walther, M. M., Merino, M. J., Choyke, P. et al.: Genetic Basis of Cancer of the Kidney: Disease-Specific Approaches to Therapy. Clin Cancer Res, 10: 6282S, 2004. 4. Linehan, W. M. and Zbar, B.: Focus on kidney cancer. Cancer Cell, 6: 223, 2004. 5. Linehan, W. M., Walther, M. M., and Zbar, B.: The genetic basis of cancer of the kidney. J Urol, 170: 2163, 2003. 6. Chen, F., Kishida, T., Yao, M., Hustad, T., Glavac, D., Dean, M. et al.: Germline mutations in the von Hippel-Lindau disease tumor suppressor gene: correlations with phenotype. Hum Mutat, 5: 66, 1995. 7. Gnarra, J. R., Tory, K., Weng, Y., Schmidt, L., Wei, M. H., Li, H. et al.: Mutations of the VHL tumour suppressor gene in renal carcinoma. Nat Genet, 7: 85, 1994. 8. Motzer, R. J., Hutson, T. E., Tomczak, P., Michaelson, M. D., Bukowski, R. M., Rixe, O. et al.: Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med, 356: 115, 2007. 9. Escudier, B., Eisen, T., Stadler, W. M., Szczylik, C., Oudard, S., Siebels, M. et al.: Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med, 356: 125, 2007. 10. Motzer, R. J., Hudes, G. R., Curti, B. D., McDermott, D. F., Escudier, B. J., Negrier, S. et al.: Phase I/II trial of temsirolimus combined with interferon alfa for advanced renal cell carcinoma. J Clin Oncol, 25: 3958, 2007. 11. Haas, N. B. and Atkins, M. B.: A New Adjuvant Trial is Born: E2805 to Study Effects of Sorafenib, Sunitinib after Nephrectomy. Kidney Cancer Journal, 4: 9, 2007. 12. Zbar, B., Tory, K., Merino, M. J., Schmidt, L., Glenn, G. M., Choyke, P. et al.: Hereditary papillary renal cell carcinoma. J Urol, 151: 561, 1994. 40
  • 47. 13. Lubensky, I. A., Schmidt, L., Zhuang, Z., Weirich, G., Pack, S., Zambrano, N. et al.: Hereditary and sporadic papillary renal carcinomas with c-met mutations share a distinct morphological phenotype. Am J Pathol, 155: 517, 1999. 14. Schmidt, L., Duh, F. M., Chen, F., Kishida, T., Glenn, G. M., Choyke, P. et al.: Germline and somatic mutations in the tyrosine kinase domain of the MET proto-oncogene in papillary renal carcinomas. Nat Gen, 16: 68, 1997. 15. Schmidt, L., Junker, K., Weirich, G., Glenn, G., Choyke, P., Lubensky, I. et al.: Two North American families with hereditary papillary renal carcinoma and identical novel mutations in the MET proto-oncogene. Cancer Res, 58: 1719, 1998. 16. Pavlovich, C. P., Walther, M. M., Eyler, R. A., Hewitt, S. M., Zbar, B., Linehan, W. M. et al.: Renal tumors in the Birt-Hogg-Dube syndrome. Am J Surg Pathol, 26: 1542, 2002. 17. Nickerson, M. L., Warren, M. B., Toro, J. R., Matrosova, V., Glenn, G. M., Turner, M. L. et al.: Mutations in a novel gene lead to kidney tumors, lung wall defects, and benign tumors of the hair follicle in patients with the Birt-Hogg-Dube syndrome. Cancer Cell, 2: 157, 2002. 18. Schmidt, L. S., Nickerson, M. L., Warren, M. B., Glenn, G. M., Toro, J. R., Merino, M. J. et al.: Germline BHD-mutation spectrum and phenotype analysis of a large cohort of families with Birt-Hogg-Dube syndrome. Am J Hum Genet, 76: 1023, 2005. 19. Grubb, R. L., III, Franks, M. E., Toro, J., Middelton, L., Choyke, L., Fowler, S. et al.: Hereditary leiomyomatosis and renal cell cancer: a syndrome associated with an aggressive form of inherited renal cancer. J Urol, 177: 2074, 2007. 20. Wei, M. H., Toure, O., Glenn, G. M., Pithukpakorn, M., Neckers, L., Stolle, C. et al.: Novel mutations in FH and expansion of the spectrum of phenotypes expressed in families with hereditary leiomyomatosis and renal cell cancer. Journal of Medical Genetics, 43: 18, 2006. 21. Merino, M. J., Torres-Cabala, C., Pinto, P., and Linehan, W. M.: The morphologic spectrum of kidney tumors in hereditary leiomyomatosis and renal cell carcinoma (HLRCC) syndrome. Am J Surg Pathol, 31: 1578, 2007. 22. Zbar, B., Glenn, G., Merino, M., Middelton, L., Peterson, J., Toro, J. et al.: Familial renal carcinoma: clinical evaluation, clinical subtypes and risk of renal carcinoma development. J Urol, 177: 461, 2007. 23. Pinto, P. A., Chien, G. W., and Shalhav, A. L.: Laparoscopic renal surgery., 2nd ed Hamilton, Ontario: B.C. Decker, Inc., 2007. 24. Williams, C. R. and Pinto, P. A.: Laparoscopic partial nephrectomy. In: Atlas of laparoscopic retroperitoneal surgery, In Press ed. Edited by Bishoff, J. T. and Kavoussi, L. R.: Milan: Springer, chapt. 2nd ed., 2017. 41
  • 48. 25. Pinto, P. A. and Jarrett, T. W.: Renal Surgery in Adults. In: Retroperitoneoscopy and Extraperitoneal Laparoscopy in Pediatric and Adult Urology. Edited by Caione, P., Kavoussi, L. R., and Micali, F.: Milan: Springer, 2003. 26. Clayman, R. V., Kavoussi, L. R., Soper, N. J., Dierks, S. M., Meretyk, S., Darcy, M. D. et al.: Laparoscopic nephrectomy: initial case report. J Urol, 146: 278, 1991. 27. Gill, I. S., Schweizer, D., Hobart, M. G., Sung, G. T., Klein, E. A., and Novick, A. C.: Retroperitoneal laparoscopic radical nephrectomy: the Cleveland clinic experience. J Urol, 163: 1665, 2000. 28. Micali, S., Caione, P., Virgili, G., Capozza, N., Scarfini, M., and Micali, F.: Retroperitoneal laparoscopic access in children using a direct vision technique. J Urol, 165: 1229, 2001. 29. Licht, M. R. and Novick, A. C.: Nephron sparing surgery for renal cell carcinoma. J Urol, 149: 1, 1993. 30. Provet, J., Tessler, A., Brown, J., Golimbu, M., Bosniak, M., and Morales, P.: Partial nephrectomy for renal cell carcinoma: indications, results and implications. J Urol, 145: 472, 1991. 31. Hafez, K. S., Fergany, A. F., and Novick, A. C.: Nephron sparing surgery for localized renal cell carcinoma: impact of tumor size on patient survival, tumor recurrence and TNM staging. J Urol, 162: 1930, 1999. 32. Ono, Y., Kinukawa, T., Hattori, R., Yamada, S., Nishiyama, N., Mizutani, K. et al.: Laparoscopic radical nephrectomy for renal cell carcinoma: a five-year experience. Urol, 53: 280, 1999. 33. Thomas, M. A., Rha, K. H., Ong, A. M., Pinto, P. A., Montgomery, R. A., Kavoussi, L. R. et al.: Optical access trocar injuries in urological laparoscopic surgery. J Urol, 170: 61, 2003. 34. Finelli, A. and Gill, I. S.: Laparoscopic partial nephrectomy: contemporary technique and results. Urologic Oncology, 22: 139, 2004. 35. Thompson, T., Ng, C. F., and Tolley, D.: Renal parenchymal hemostatic aids: glues and things. Current Opinions in Urology, 13: 209, 2003. 36. Tsivian, A. and Sidi, A. A.: A simple and reliable hemostatic technique during partial nephrectomy. Urol, 63: 976, 2004. 37. Allaf, M. E., Bhayani, S. B., Rogers, C., Varkarakis, I., Link, R. E., Inagaki, T. et al.: Laparoscopic partial nephrectomy: evaluation of long-term oncological outcome. J Urol, 172: 871, 2004. 42