return to agenda

Biomarkers in Cancer Therapeutics

 

Objectives

·        Assess the use of imaging modalities as correlates to physiologic markers

·        Evaluate various approaches used to identify cellular markers of disease state, progression of disease, and therapeutic competence (efficacy)

·        Determine research needs for the advancement of marker identification useful in the identification of early cancer detection and cancer therapeutic drug targets

·        Identify other technological approaches that will provide opportunities for developing more and better biomarkers to measure therapeutic efficacy

·        Examine the roles of industry, academia, and government in the development of toxicity biomarkers

·        Determine the current barriers impeding the development of biomarkers for cancer therapeutics

 

Agenda

Moderators:   Albert F. LoBuglio, M.D., University of Alabama, Birmingham

James M. Pluda, M.D., National Cancer Institute

S. Percy Ivy, M.D., National Cancer Institute

 

Introduction and Overview

Albert F. LoBuglio, M.D.

Biomarkers and the Impact on Drug Development

Biomarker Analysis in Human Carcinomas Subsequent to Epidermal Growth Factor-Receptor Blockade Therapy Alone and in Combination With Cytotoxic Agents

Robert Radinsky, Ph.D., University of Texas M.D. Anderson Cancer Center

Biochemical Endpoints in Mechanism-Based Clinical Cancer Trials

James K.V. Willson, M.D., University Hospitals of Cleveland and Case Western Reserve University

Practical Issues in Current Drug Development:  The Gross Philadelphia Chromosome as a Chronic Myelogenous Leukemia Surrogate Endpoint; Progression-Free Survival as an Endpoint for Tumoristatic Therapies

Robert J. Spiegel, M.D., Schering-Plough Research Institute

HER2 Testing:  Use of a Biomarker To Select Therapy

Susan D. Hellmann, M.D., M.P.H., Genentech, Inc.

 

Biomarker Application in the Assessment of Cellular Mechanisms

Biomarkers and Angiogenesis—A Tabula Rasa

Thomas Boehm, Ph.D., Children’s Hospital and Harvard Medical School

Prognostic Markers

Donald Berry, Ph.D., Duke University

 

Biomarkers in Cancer Therapeutics (continued)

Technology Interface With Pathophysiology

Radiolabeled Probes as a Tool for the Assessment of Receptor Expression or Vascularity

Albert F. LoBuglio, M.D.

Assessing the Effects of Antiangiogenic Therapy on Tumor Vasculature and Metabolism Using Functional Imaging

Steven K. Libutti, M.D., National Cancer Institute

Open Discussion

Summary

James M. Pluda, M.D.

Albert F. LoBuglio, M.D.

S. Percy Ivy, M.D.

Biomarker Analysis in Human Carcinomas Subsequent to Epidermal Growth Factor‑Receptor Blockade Therapy Alone and in Combination With

Cytotoxic Agents

C.J. Bruns, M.D.; P. Perrotte, M.D.; T. Matsumoto, M.D., Ph.D.; C.P.N. Dinney, M.D.; and Robert Radinsky, Ph.D.

The molecular mechanisms specific regulate the metastasis of tumor cells to specific organs are diverse and both tumor and organ specific.  Data will be presented that demonstrate that the organ microenvironment can influence the pattern of gene expression and the biologic phenotype of metastatic tumor cells, including regulation of cellular survival, angiogenesis, and growth at the organ‑specific metastatic site.  Insight into the molecular mechanisms regulating this process as well as a better understanding of the interaction between the metastatic cell and the organ‑specific microenvironment provides a foundation for the design of new therapeutic approaches.  The purpose of this study was to determine whether epidermal growth factor receptor (EGF‑R) signaling regulates, in part, human bladder and pancreatric carcinoma cell proliferation, invasion, or angiogenesis.  EGF‑R overexpression correlates with bladder (TCC) and pancreatic carcinoma (PC) progression.  We evaluated whether EGF‑R blockade by use of (1) dominant negative mutant EGF‑Rs, (2) EGF‑R‑specific tyrosine kinase inhibitors, or (3) neutralizing anti‑EGF‑R antisera (C225) has therapeutic benefits against high‑grade TCC and PC growing orthopically in nude mice.  In vitro treatment of each cell type with EGF‑R blockade therapies resulted in inhibition of EGF‑R‑specific phosphorylation as measured by Western blotting and maximal 50 percent cytostasis.  A decrease was observed in expression of the angiogenic factors vascular endothelial growth factor, IL‑8, and bFGF and the matrix metalloproteinase 9 protease by the treated cells at both mRNA and protein levels in a dose‑dependent manner (p<0.005).  In contrast, cell cycle‑related proteins such as p16, p21, and CDK2 were not downregulated, although the cyclin‑dependent kinase inhibitor p27 was elevated after treatment.  Systemic therapy of established TCC and PC tumors with EGF‑R blockade therapies alone or in combination with Taxol or gencitabine, respectively, resulted in growth inhibition, tumor regression, and abrogation of metastasis (p<0.0005).  Therapy conferred a significant survival advantage to the treated mice (p=0.0001).  The expression of proliferating cell nuclear antigen, Rb, MMP9, VEGF, IL‑8, and bFGF (protein and mRNA) levels was significantly reduced in treated versus control tumors (p=0.001), whereas p27 levels were induced to high levels.  The inhibition of the angiogenic peptides resulted in the subsequent involution of the tumor neovascularization as determined by microvessel density (p<0.005), contributing in part to an increased tumor cell apoptotic index.  These experiments indicate that therapeutic strategies targeting EGF‑R signaling have a significant antitumor effect, mediated in part by the inhibition of cellular proliferation, invasion, and angiogenesis that leads to apoptosis and tumor regression.  Ongoing studies are analyzing the identical biomarkers in patient specimens subsequent to treatment with EGF‑R blockade therapies in combination with cytotoxic drugs.  Collectively, these data support the hypothesis that the microenvironment of different organs can influence the biological behavior of tumor cells during the metastatic process and provide a therapeutic basis for interfering with metastasis by downregulation of receptor number or function.

 

Biochemical Endpoints in Mechanism‑Based Clinical Cancer Trials

James K.V. Willson, M.D.

Our group has conducted a series of mechanism‑based phase I clinical cancer trials in which one endpoint of the trial was the modulation of a molecular target.  A procedure for sequential computerized tomography‑guided biopsies of solid tumors was developed to investigate the modulation of a biochemical target in tumor tissues.  In one phase I trial, we used this approach to determine the dose and schedule of a novel modulator of DNA alkyltransferase activity in tumor tissues.  In this trial, 28 patients completed sequential tumor biopsies which proved informative for biochemical studies of alkyltransferase.  The drug level required to deplete the enzyme in the tumors was tenfold higher than the drug area under the curve required to deplete alkyltransferase activity in peripheral blood mononuclear cells.  This experience demonstrates the feasibility of using molecular endpoints in the development of cancer therapeutics. This experience also demonstrates the limitation of studying a surrogate tissue and the importance of investigating biochemical endpoints in the relevant target tissue.

 

Key References

Spiro TP, Willson JKV, Haaga J, Hoppel CL, Liu L, Majka S, Gerson SL. O6‑benzylguanine DNA alkyltransferase directed DNA repair. Proc Am Soc Clin Oncol 1996;15:178(A366).

Willson JKV, Haaga JR, Trey JE, Stellato TA, Gordon NH, Gerson SL. Modulation of O6alkylguanine alkyltransferase directed DNA repair in metastatic colon cancers. J Clin Oncol 1995;13:2301‑2308.

 

Practical Issues in Current Drug Development:  The Gross Philadelphia Chromosome as a Chronic Myelogenous Leukemia Surrogate Endpoint; Progression‑Free Survival as an Endpoint for Tumoristatic Therapies

Robert J. Spiegel, M.D.

The question to be addressed is, Are there now instances where outcomes short of survival should be accepted as appropriate surrogates for new therapies in the treatment of cancer?  In particular, Does the new genetic understanding of the basis of certain malignant transformations allow demonstrations of effect on tumor genetics to serve as adequate surrogates of new therapeutic's efficacy?  In chronic myelogenous leukemia, the gross Philadelphia chromosome (Ph1) aberration has been recognized for decades as a pathognemonic marker of this disease.  With the introduction of interferon and bone marrow transplantation, for the first time some patients were demonstrated to lose detectable Ph1 following treatment.  A number of studies have now confirmed that those patients who lose Ph1 can expect durable long‑term responses.  The question arises, Can the demonstration of loss of Ph1 and normalization of hematologic values for some X period of time be considered a suitable surrogate of treatment efficacy and preclude the necessity of following cohorts of hundreds of patients over 3 to 5 years to demonstrate survival benefit?  This model raises interesting questions for other therapies in the future, which may be able to eliminate specific genetic defects that are easily tracked.  A larger and related question involves the recent introduction into the clinic of new therapies that are expected to produce tumoristatic rather than tumoricidal effects.  Some current approaches to gene therapy such as replacement of tumor suppressor genes (p53) or reversal of oncogenic gene stimuli (farnesyl protein transferase inhibitors in RAS‑positive tumors) may produce successful control of tumor growth but without bystander effect and without dramatic conventional tumor responses.  These approaches to cancer therapy may well produce control of existent tumor but not elimination.  Demonstration of clinical benefit in these circumstances may take hundreds of patients followed for many years, and this produces serious challenges to the normal development process where early readout is necessary to optimize drug formulations or determine a positive decision to invest further resources.  A discussion of specific therapeutic approaches and specific disease settings will be made.

 

Key References

Anon. Interferon alfa‑2a as compared with conventional chemotherapy for the treatment of chronic myeloid leukemia. The Italian Cooperative Study Group on Chronic Myeloid Leukemia. N Engl J Med 1994;114:820‑825.

Guilhot F, Chastang C, Michallet M, Guerci A, Harousseau JL, Maloisel F, Bouabdallah R, Guyotat D, Cheron N, Nicolini F, Abgrall JF, Tanzer J. Interferon alpha‑2b combined with cytarabine versus interferon alone in chronic myelogenous leukemia. French Chronic Myeloid Leukemia Study Groups. N Engl J Med 1997;337:223‑229.

Kantarjian HM, Smith TL, O'Brien S, Beran M, Pierce S, Talpaz M. Prolonged survival in chronic myelogenous leukemia after cytogenetic response to interferon‑alpha therapy. The Leukemia Service. Ann Intern Med 1995;122:254‑261.

Ohnishi K, Ohno R, Tomonaga M, Kamada N, Onozawa K, Kuramoto A, Dohy H, Mizoguchi H, Miyawaki S, Tsubaki K, et al. A randomized trial comparing interferon-alpha with busulfan for newly diagnosed chronic myelogenous leukemia in chronic phase. Blood 1995;86:906‑916.

Talpaz M, Kantarjian H, Kurzrock R, Trujillo JM, Gutterman JU. Interferon‑alpha produces sustained cytogenetic responses in chronic myelogenous leukemia. Philadelphia chromosome‑positive patients. Ann Intern Med 1991;114:532‑538.

 

HER2 Testing:  Use of a Biomarker to Select Therapy

Susan D. Hellmann, M.D., M.P.H.

The use of biomarkers to select therapy has been a frequent topic for theoretical discussion.  Herceptin, a recently approved humanized monoclonal antibody, is the first drug whose use is specifically based on the presence of a biomarker, the HER2/neu oncogene.  The challenges associated with the practical implications of using a biomarker to select therapy will be discussed.

 

Biomarkers and Angiogenesis—A Tabula Rasa

Thomas Boehm, Ph.D. 

The field of angiogenesis research and particularly the application of the antiangiogenesis concept as a potential way to treat cancer were pioneered by Judah Folkman during the past 30 years.  During the past 5 years, several potent or less potent molecules have been found to be capable of controlling certain aspects of angiogenesis and thereby of blocking tumor growth.  Nevertheless, only a handful publications can be found on biomarker and angiogenesis.  In addition, these publications describe molecules secreted from tumor cells to stimulate endothelial cells (bFGF and VEGF).  In other words, these proteins are not real markers for angiogenesis but for the presence of tumor cells.  It is now generally accepted that tumor cells need to establish an adequate capillary network to expand.  This means that endothelial cells will undergo phenotypical changes, because they have to migrate, proliferate, degrade, and remodel the matrix to accomplish the task of forming a functional unit supplying the tumor cells with oxygen and nutrients.  If these phenotypical changes can be measured and quantified, we could detect cancer earlier or follow antiangiogenic therapy.  Before this can be accomplished, these endothelial cell‑specific molecules lost or overexpressed during tumor growth must be identified.  A possible approach to isolate real endothelial cell marker will be presented.

 

Key References

Dirix LY, Vermeulen PB, Pawinski A, Prove A, Benoy I, De Pooter C, Martin M, Van Oosterom AT. Elevated levels of the angiogenic cytokines basic fibroblast growth factor and vascular endothelial growth factor in sera of patients. Br J Cancer 1997;76(2):238‑243.

Jinno K, Tanimizu M, Hyodo I, Nishikawa Y, Hosokawa Y, Doi T, Endo H, Yamashita T, Okada Y. Circulating vascular endothelial growth factor (VEGF) is a possible tumor marker for metastasis in human hepatocellular carcinoma. J Gastroenterol 1998;33(3):376‑382.

Kumar H, Heer K, Lee PW, Duthie GS, MacDonald AW, Greenman J, Kerin MJ, Monson JR. Preoperative serum vascular endothelial growth factor can predict stage in colorectal cancer. Clin Cancer Res 1998;4(5):1279‑1285.

Landriscina M, Cassano A, Ratto C, Longo R, Ippoliti M, Palazzotti B, Crucitti F, Barone C. Quantitative analysis of basic fibroblast growth factor and vascular endothelial growth factor in human colorectal cancer. Br J Cancer 1998;78(6):765‑770.

Moses MA, Wiederschain D, Loughlin KR, Zurakowski D, Lamb CC, Freeman MR. Increased incidence of matrix metalloproteinases in urine of cancer patients. Cancer Res 1998;58(7):1395‑1399.

Nguyen M, Watanabe H, Budson AE, Richie JP, Hayes DF, Folkman J. Elevated levels of an angiogenic peptide, basic fibroblast growth factor, in the urine of patients with a wide spectrum of cancer. J Natl Cancer Inst 1994;86(5):356‑361.

 

 

Prognostic Markers

Donald Berry, Ph.D.

Prognostic markers are of little clinical interest unless they have therapeutic implications. Despite some attitudes to the contrary, it may not be appropriate to use the most intensive and most toxic therapies with patients having the poorest prognosis.  But assessing interactions between therapy and biomarkers is difficult and requires very large numbers of patients.  Such assessment is made even more difficult because the typical experiment involves assessing numerous markers.  A result is that the false‑positive rate increases, giving rise to biomarkers that are flashes in the pan.  These and related problems in the case of HER‑2/neu and p53 and their possible interaction with doxorubicin in node‑positive breast cancer will be discussed as well as the general notion of using surrogate markers in clinical research from the Bayesian perspective.


Radiolabeled Probes as a Tool for the Assessment of Receptor Expression

or Vascularity

Albert F. LoBuglio, M.D.

A recent phase I clinical trial utilizing a humanized anti‑Vitronectin receptor provided an opportunity for our group to initiate attempts at measuring antibody binding or blockade of this angiogenesis‑dependent endothelial cell receptor.  We proposed that it may bind available Vitronectin receptors and that administration of radiolabeled tracer doses might be used to image tumor vascular bed and when administered postinfusion of a therapeutic dose, might demonstrate blockade of the receptor.  This initial attempt was only partially successful and suggested that the antibody’s binding affinity was too low, and particularly, its “off‑rate” was too rapid.  Evidence for this hypothesis was then presented by Viti and colleagues who showed that single‑chain antibodies directed to an alternative intravascular target were only able to image the tumor vascular bed if their off‑rate was very slow.  Such high‑affinity and low off‑rate versions of the anti‑Vitronectin receptor antibody have been described.  The generalization of this concept (i.e., use of a probe carrying an isotope or other imaging agent as a means of delineating the tumor vascular bed) might well be applicable to magnetic resonance imaging and positron emission tomography scanning as a means of estimating “volume” or “extent” of vascular bed per volume of tumor.

 

Key References

Brooks PC, Clark RA, Cheresh DA. Requirement of vascular integrin alpha v beta 3 for angiogenesis. Science 1994;264(5158):569‑571.

Viti F, Tarli L, Giovannoni L, Zardi L, Neri D. Increased binding affinity and valence of recombinant antibody fragments lead to improved targeting of tumoral angiogenesis. Cancer Res 1999;59(2):347‑352.

 

Assessing the Effects of Antiangiogenic Therapy on Tumor Vasculature and Metabolism Using Functional Imaging

Steven K. Libutti, M.D.

As new anticancer agents are developed that target the tumor neovasculature, novel approaches to measuring responses are needed.  Functional imaging has been suggested as a means of detecting changes in tumor blood flow and metabolism.   Several challenges confront the development of these new technolgies.  We must identify noninvasive methods to quantify changes in tumor vasculature, and we must validate these methods as a means of quantifying the effects of antivascular agents.  We must also be able to control for variability of repeated measures.  Finally, we must be able to coregister images with traditional scans to follow the responses of particular lesions over time.  We have identified several advanced imaging modalities that we believe have the potential to address these challenges:  positron emission tomography (PET) utilizing radiolabeled glucose (18FDG), radiolabeled carbon monoxide (11CO), radiolabled water (H2O15), and dynamic magnetic resonance imaging (MRI).  PET‑FDG relies on the fact that malignant lesions have elevated glycolysis, and therefore there is increased uptake of [18‑F] fluorodeoxyglucose.  Conversion of 18FDG to 18FDG‑6‑phosphate occurs, which traps the radioisotope in neoplastic tissues.  Our hypothesis is that changes in tumor vascularity in response to agents may alter glucose metabolism and therefore change the amount of uptake of 18FDG.  PET‑CO is designed to assist in the quantification of tumor blood volume.  11CO is administered by inhalation, binds tightly to red blood cells, and has a short half‑life (20 minutes).  Red blood cell volume can be quantified as can changes in blood volume over time.  PET‑H2O allows for the measurement of tumor blood flow.  H2O15  is delivered intravenously and is rapidly cleared (half‑life < 5 minutes).  Short scanning times can be used, and flow in tumor vessels can be standardized to major vessels and to arterial blood samples.  The technique has been validated in myocardial and cerebral blood flow studies.  Dynamic MRI relies on the rapid (more than 1 minute) administration of a gadolinium‑based contrast agent.  Ultrafast analysis of signal intensity and patterns of contrast uptake within tumors can correlate with microvessel density.  Pharmacokinetic modeling of gadolinium clearance—as well as measurements such as the rate of enhancement, time to peak enhancement, clearance rate, and area under the curve—can be performed.  Recent advances in MRI utilizing gradient‑recalled echo and inhaled carbogen may enhance these measurements.  Our current clinical trial is designed to evaluate these various imaging modalities in patients currently being treated with antiangiogenic agents (anti‑VEGF, thalidomide, isolated hepatic perfusion).  Pretreatment imaging with CT, PET, and dynamic MRI is followed by interval imaging while on therapy.  Correlation with tumor biopsy is performed when available.