You are leaving Medscape Education
Cancel Continue
Log in to save activities Your saved activities will show here so that you can easily access them whenever you're ready. Log in here CME & Education Log in to keep track of your credits.
 

CME

Signal Transduction Inhibition: Current Knowledge, Pathways, and Experience in Cancer Therapy

  • Authors: Faculty: Eric Rowinsky, MD; Mary-Ann Bjornsti, PhD; Sandrine Faivre, MD, PhD; Peter Houghton, PhD; Funda Meric-Bernstam, MD; Gordon Mills, MD, PhD; Eric Raymond, MD, PhD
  • THIS ACTIVITY HAS EXPIRED
Start Activity


Target Audience and Goal Statement

The primary target audience for this activity is medical oncologists and hematologists/oncologists.

Upon completion of this activity, participants will be able to:

  1. Explain signal transduction pathways involving apoptosis and cell cycle inhibition in cancer.
  2. Evaluate mechanisms leading to the pathogenesis of solid tumors.
  3. Discuss current paradigms in the treatment of cancer.
  4. Describe the role of inhibition of signal transduction as a therapeutic modality in solid tumors.


Disclosures

The content and views presented in this educational activity are those of the authors and do not necessarily reflect those of the Dannemiller Memorial Educational Foundation, inRx Medical Education, or Wyeth Pharmaceuticals. This material has been prepared based upon a review of multiple sources of information, but it is not exhaustive of the subject matter. Therefore, healthcare professionals and other individuals should review and consider other publications and materials on the subject matter before relying solely upon the information contained within this educational activity.

Discussions of imatinib mesylate for CML and GIST, cetuximab, gefitinib, trastuzumab, and bevacizumab are FDA-approved uses. All other discussions may involve off-label use or products not approved by the FDA.


Author(s)

  • Eric Rowinsky, MD

    Program Leader, Experimental Therapeutics, San Antonio Cancer Institute, San Antonio, Texas.

    Disclosures

    Disclosure: Research Grant: Wyeth Pharmaceuticals, ARIAD Pharmaceuticals, Novartis Pharmaceuticals; Advisory Board: Wyeth Pharmaceuticals, ARIAD Pharmaceuticals, Novartis Pharmaceuticals.

  • Eric Raymond, MD, PhD

    Head, Translational and Clinical Pharmacology, Saint-Louis Hospital, Paris, France.

    Disclosures

    Disclosure: Dr. Raymond has nothing to disclose.

  • Gordon Mills, MD, PhD

    Professor and Chairman, Molecular Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas.

    Disclosures

    Disclosure: Grant: Celgene, Echelon Corporation, Eli Lilly and Company, Kinetek Pharmaceuticals; Consultant: Abbot Laboratories, Odyssey Thera, Celgene, Echelon Corporation, Eli Lilly and Company, Kinetek Pharmaceuticals, Pinto Ventures; Stockholder: Maxum; Research Support: Celgene, Echelon Corporation, Eli Lilly and Company, Kinetek Pharmaceuticals; Honorarium: Abbott Laboratories, Celgene, Echelon Corporation, Eli Lilly and Company, Kinetek Pharmaceuticals, Odyssey Thera, Pinto Ventures.

  • Funda Meric-Bernstam, MD

    Assistant Professor, Surgical Oncology, M.D. Anderson Cancer Center, Houston, Texas.

    Disclosures

    Disclosure: Dr. Meric-Bernstam has nothing to disclose.

  • Peter Houghton, PhD

    Chair, Molecular Pharmacology, St. Jude Children's Research Hospital Memphis, Tennessee.

    Disclosures

    Disclosure: Research Grant: Wyeth Pharmaceuticals.

  • Mary-Ann Bjornsti, PhD

    Associate Member, Department of Molecular Pharmacology, St. Jude Children's Hospital Memphis, Tennessee.

    Disclosures

    Disclosure: Dr. Bjornsti has nothing to disclose.

  • Sandrine Faivre, MD, PhD

    Assistant Professor, Medicine, Gustave-Roussy Institute, Villejuif, France.

    Disclosures

    Disclosure: Dr. Faivre has nothing to disclose.


Accreditation Statements

    For Physicians

  • Sponsored by Dannemiller Memorial Educational Foundation

    The Dannemiller Memorial Educational Foundation is accredited by the Accreditation Council for Continuing Medical Education to provide continuing medical education for physicians.

    The Dannemiller Memorial Educational Foundation designates this educational activity for a maximum of 3.0 category 1 credits toward the AMA Physician's Recognition Award. Each physician should claim only those credits that he/she actually spent in the activity.

    Contact This Provider

For questions regarding the content of this activity, contact the accredited provider for this CME/CE activity noted above. For technical assistance, contact [email protected]


Instructions for Participation and Credit

There are no fees for participating in or receiving credit for this online educational activity. For information on applicability and acceptance of continuing education credit for this activity, please consult your professional licensing board.

This activity is designed to be completed within the time designated on the title page; physicians should claim only those credits that reflect the time actually spent in the activity. To successfully earn credit, participants must complete the activity online during the valid credit period that is noted on the title page.

Follow these steps to earn CME/CE credit:

  1. Read the target audience, learning objectives, and author disclosures.
  2. Study the educational content online or printed out.
  3. Online, choose the best answer to each test question. To receive a certificate, you must receive a passing score as designated at the top of the test. Medscape encourages you to complete the Activity Evaluation to provide feedback for future programming.

You may now view or print the certificate from your CME/CE Tracker. You may print the certificate but you cannot alter it. Credits will be tallied in your CME/CE Tracker and archived for 5 years; at any point within this time period you can print out the tally as well as the certificates by accessing "Edit Your Profile" at the top of your Medscape homepage.

The credit that you receive is based on your user profile.

CME

Signal Transduction Inhibition: Current Knowledge, Pathways, and Experience in Cancer Therapy

Authors: Faculty: Eric Rowinsky, MD; Mary-Ann Bjornsti, PhD; Sandrine Faivre, MD, PhD; Peter Houghton, PhD; Funda Meric-Bernstam, MD; Gordon Mills, MD, PhD; Eric Raymond, MD, PhDFaculty and Disclosures
THIS ACTIVITY HAS EXPIRED

processing....

The Developing Diversity of Targeted Therapy in Oncology, Presented by by Gordon Mills, MD, PhD; Funda Meric-Bernstam, MD

Introduction

The relatively narrow therapeutic window characteristic of the vast majority of traditional chemotherapeutics has led to pursuit of targeted therapies with the hope of providing both improved efficacy and reduced toxicity. These targeted therapies use agents that impact specific factors or cellular pathways that have been shown to have abnormal activity in neoplastic cells and that drive cancer progression. The diversity of potential targets is currently most impressive, with an astounding number of new possibilities emerging. However, translation of these targets into useful approaches for clinical drug development has provided both exciting successes and disappointing failures. Some of the most attractive targets for molecular-targeted strategies for the treatment of cancer include the family of receptor and non-receptor tyrosine kinases (TKs) and more specific areas of the growth factors and epidermal growth factor receptor (EGFR) inhibitors; the Ras/Raf/MAPK pathway; thephosphatidylinositol-3 kinase (PI3K)/Akt/PTEN pathway, gene expression modification with antisense oligonucleotides and RNAi; and endothelial cell and angiogenesis-associated factors such as the vascular endothelial cell growth factor (VEGF).[1-3]

Although some of these approaches have produced clinically useful agents, primarily due to the nature of the abnormality targeted, other approaches have not been as successful because of the redundancies and overlapping nature of complex signal transduction pathways. In many cases, although the aberration is present in cancer cells, it does not drive tumor behavior. Indeed, one of the most significant challenges is to distinguish molecular events that drive tumor behavior from epiphenomena that arise secondarily to an important event or due to the inherent genetic instability in tumors.[4] Although single-agent therapy remains a goal, experience with conventional cytotoxic therapy and with experimental agents suggests that combining these new, targeted agents with either conventional therapies or one another is likely to provide the best efficacy. Indeed, given the complex crosstalk between signaling pathways, it is likely that it will be necessary to hit severalcritical signaling nodes to attain optimal outcomes. However, as combinations of targeted therapeutics may be more effective, they may also be more toxic, a process that can be quantitated only in patient studies.[4]

Signal Transduction Kinases

  • Kinases are enzymes that regulate the function of other proteins/enzymes and other target molecules. Tyrosine kinases accomplish signal transduction by transferring an activating gamma phosphate group from ATP to a tyrosine residue on the substrate molecule, usually another protein. Serine and threonine kinases are a separate group of transduction enzymes that target serine and threonine residues rather than tyrosine residues on substrate molecules. In addition to proteins, a number of other intracellular signaling molecules, such as membrane lipids or intracellular inositol rings, are regulated by their phosphorylation status through the action of specific kinases. Phosphatases remove phosphates on the substrate molecules and thus oppose kinase activity and inhibit the signal transfer. These 2 enzyme classes act as switches; they activate and deactivate pathways according to the signals received by the cell. Although in general, phosphorylation events positively regulatesignaling, there are circumstances in which phosphorylation inhibits function. So depending on the cell system and the target, kinases and phosphatases can be either positive or negative regulators. Cellular signaling frequently involves phosphorylation of all 3 amino acid residues (tyrosine, threonine, and serine), sometimes at multiple sites on one protein, or of other signaling molecules such as lipids and on multiple components in a pathway, suggesting that multiple kinases and kinase classes function collaboratively to orchestrate the final outcome of signal transduction. Figure 1 is a simplified example of a model signal transduction pathway. An extracellular signal activates a membrane-bound receptor, such as EGFR, which forms either homodimers with another EGFR or heterodimers with other family members to initiate the signal relay across the cytoplasm, culminating in changes of gene transcription in the nucleus and subsequent regulation of important cellular functions such ascell proliferation, migration, or survival.[5]

    Some kinases are membrane bound, and others reside in the cytoplasm. Thus, development of agents to modify kinase activities has included antibodies targeted to the extracellular portion of the membrane-bound kinase, eg, trastuzumab, as well as small molecules that act in the cytoplasm, eg, imatinib mesylate.[1,2] Inhibition of intracellular TK activity has been achieved using small molecules whose structures are based on the structure of ATP. This characteristic allows these molecules to affect not only tumors that overexpress TK activity, but also those with normal TK expression or mutated, constitutively activated forms of TK. These competitive inhibitors of the ATP-binding site on the kinase can be specific for individual kinases or have a broader spectrum inhibiting multiple kinases, such as the effects of imatinib mesylate on Abl, Kit (CD 117), and platelet-derived growth factor (PDGFR), and have the clinical benefit of oral bioavailability.[6]

  • Figure

    Figure 1.

    Simplified signal transduction cascade through binding of a ligand to the membrane receptor to final effects on gene expression in the nucleus.[5]

    (Enlarge Slide)

The Bcr-Abl Tyrosine Kinase and Inhibition by Imatinib

  • Imatinib mesylate (STI571) was the first agent in the class of small-molecule inhibitors of TK activity proven to be clinically useful, establishing intracellular kinases as "druggable" targets. Imatinib was designed to inhibit the constitutively activated cytoplasmic Bcr-Abl kinase created by the Philadelphia chromosome abnormality in chronic myeloid leukemia (CML). Downstream proliferative signal transduction and the inhibition of apoptosis were thus prevented (Figure 2).[7] The agent also inhibits 2 other TKs, Kit and PDGFR. Because of this activity, imatinib is also used for the treatment of Kit-positive unresectable or metastatic gastrointestinal stromal tumors (GISTs), PDGFR-driven hypereosinophilic syndrome, and dermatofibrosarcoma protuberans.[8-10] Indeed, imatinib mesylate appears to demonstrate activity in cells with mutations or rearrangement in Abl, Kit, or PDGFR, but to have limited activity in tumors without genetic aberrations in these targets.This suggests that the presence of a genetic aberration provides an indication that the target is indeed driving tumor progression. The high rates of response to imatinib in these diseases are exciting; however, they may not represent the results to be expected in most epithelial tumors. This set of tumors may represent early disease in which the tumors have not yet acquired the multiple genetic aberrations that are found in most epithelial tumors.

    Studies with GIST have proven particularly informative. Patients with perimembrane-activating mutations in Kit have shown high-frequency, durable responses. Patients with mutations at other sites in Kit have shown both a lower frequency of response and short duration of responses.[11] This observation suggests that, while the presence of mutations may signal tumors driven by a particular enzyme, not all mutations are created equal. Even more intriguing is the observation that positron emission tomography (PET) imaging of FDG-glucose uptake can identify patients destined to respond to therapy early in the course of treatment.[9] Indeed, a decrease in FDG-glucose uptake within the first week of therapy indicates eventual responsiveness, which may not be evidenced using other technologies, including computed axial tomography (CAT) scans, for several months. Strikingly, a failure to normalize FDG-glucose uptake indicates that a patient is unlikely to respond totherapy. Thus, molecular imaging can predict responsiveness and allow persistence of therapy or early change to more effective approaches. Every effort must be made to identify molecular imaging approaches applicable to the plethora of targeted therapeutics under development.

  • Figure

    Figure 2.

    Inhibition of Bcr-Abl by imatinib. The aberrant phosphorylation and activation of downstream signal transduction by the Bcr-Abl fusion protein is inhibited by the competitive binding of imatinib in the ATP-binding site.[7]

    (Enlarge Slide)

The ErbB Receptor Tyrosine Kinases

  • The ErbB family of receptor tyrosine kinases (RTKs) encompasses 4 closely related transmembrane receptors: erbB-1 (EGFR or HER-1), erbB-2 (HER-2 or neu), erbB-3 (HER-3), and erbB-4 (HER-4).[12,13] These receptors exhibit similar structures with 3 distinct regions: an extracellular ligand-binding domain, a transmembrane region, and an intracellular region with tyrosine kinase activity. Receptor activation occurs after a ligand binds to the extracellular domain, which triggers dimer formation. The dimers can be either homodimers or heterodimers of the various erbB proteins. Indeed, erbB-2 does not appear to have a specific ligand, but rather functions as a dimerization partner for other family members. When overexpressed, it appears to form constitutively active homodimers. Dimerization causes activation of the intracellular kinase via autophosphorylation of tyrosine moieties in its intracellular domain and transmission of downstream signaling molecules.Intriguingly, erbB-3 appears to be kinase inactive and instead functions as a linker molecule, altering or amplifying the spectrum of molecules phosphorylated by members of this family. The potential for dimerization with another erbB family member or self-association magnifies or redirects the downstream impact of elevated or aberrant erbB-RTK function.

  •  
  • Studies have shown that abnormal EGFR signaling can result from increased EGFR expression, mutation of EGFR, or decreased activity of opposing phosphatases. Downstream pathways activated by EGFR signaling include the Ras/Raf/MAPK cascade, which is implicated in proliferation, migration, and differentiation. EGFR also signals through the PI3K/Akt pathway, which is involved in proliferation and anti-apoptosis/survival.[1,2,14] These membrane-bound receptor kinases offer 2 opportunities for regulation of signal transduction: 1) through the extracellular domain, especially with monoclonal antibody blockade; and 2) through small-molecule inhibition of the intracellular kinase activity (Figure 3).[13]

    The overexpression of erbB-2/HER-2 in many cancers and, in particular, in approximately 25% of breast cancers, is well established. It is also an indicator of poor prognosis for breast, colorectal, gastric, non-small cell lung, ovarian, and prostate cancers and melanoma.1 erbB-2/HER-2 is the target for monoclonal antibody therapy with trastuzumab for patients with metastatic breast cancer overexpressing erbB-2/HER-2.[6,15,16] Success of trastuzumab, a monoclonal antibody therapy directed toward an erbB RTK, stimulated interest in antibody-targeted therapy for EGFR.

    EGFR overexpression and/or abnormal EGFR-TK activity is associated with many human malignancies, including head and neck, colorectal, non-small cell lung, gastric, pancreatic, ovarian, breast, and prostate cancers and glioma.[1,2] In these malignancies, overexpression of EGFR has been shown to be an indicator of advanced disease and poor prognosis.[14] Blockade of the receptor with mAb 225 (C225 or cetuximab) or ABX-EGF competitively inhibits the activation of the EGFR-TK.[17] Cetuximab has recently been approved for the treatment of advanced colorectal cancer in patients whose tumors express EGFR. In a study of cetuximab combined with irinotecan, tumor shrinkage was observed in 22.9% of patients and tumor progression was delayed by approximately 4.1 months. Monotherapy with cetuximab showed a tumor response rate of 10.8% and a tumor progression delay of 1.5 months.[18]

  • Figure

    Figure 3.

    EGFR signal transduction and EGFR domain targeting by monoclonal antibodies and small-molecule tyrosine kinase inhibitors.[2]

    (Enlarge Slide)
  • A number of small-molecule inhibitors of the EGFR-TK have been approved for cancer treatment, and others are in various stages of clinical development.[1,6] These agents are classed by their selectivity (monofunctional, erbB-1/EGFR-specific versus multifunctional, pan-erbB activity) and the reversibility of their inhibition (Table 1). Generally, these small molecules block the enzyme activity of transmembrane kinase, autophosphorylation, and subsequent signal transduction. They are given orally and generally show only mild to moderate toxicities, predominantly a class-associated acneiform skin rash in 50% to 70% of patients and diarrhea in approximately 40% of patients due to the normal presence of high levels of EGFR in these sites.[1,6] Although somewhat controversial, several studies demonstrate a correlation between these side effects and patient outcomes. This has been posited to be due to delivering a "biologically relevant dose" or, alternatively, torepresent an indication of immunological competence. Of these agents, gefitinib and erlotinib are the most advanced in clinical trials.

    In preclinical studies, gefitinib has shown antitumor activity in breast, lung, and ovarian cancers. Gefitinib has been approved in the United States for monotherapy of patients with advanced non-small cell lung cancer (NSCLC) whose cancer has progressed after treatment with platinum-based and docetaxel chemotherapies. In a study of patients with NSCLC (N = 216), including 142 with refractory disease, the response rate (at least 50% tumor shrinkage lasting for 1 month) was about 10%.25 For some patients, the response was more durable, with a median duration of 7 months. Since the use of the agent is third line (when no other viable therapy exists), the low response rate was thought to confer some clinical benefit. However, in 2 controlled, randomized clinical trials (N = 2130) of first-line treatment for NSCLC, gefitinib failed to confer any benefit when added to standard, platinum-based therapy.[26,27] Similar results have been seen with other chemotherapy agents suchas gemcitabine and paclitaxel.[28] As a result, gefitinib is not indicated for use in this setting. Moreover, an approximate 1% occurrence of interstitial lung disease has been reported, with approximately 30% of these occurrences resulting in deaths.19 In part due to the observation that cell lines expressing erbB-2 are sensitive to gefitinib and that in combination with trastuzumab, gefitinib exhibits a synergistic inhibition in cells lines co-expressing erbB-1 and erbB-2, this agent has been studied in patients with advanced breast cancer.[2,29] Although the participating patients (N = 63) were not screened for EGFR, 43% had tumors that were estrogen-receptor positive and 27% overexpressed erbB-2. The objective response was low in this heavily pretreated population, and only 14.3% of patients achieved a partial response or stable disease that endured for up to 6 months. Gefitinib is currently being studied in a wide diversity of clinical trials, includingmonotherapy for refractory ovarian cancer and combination therapy for metastatic breast cancer and bladder cancer.[30]

    Recent studies have demonstrated that individuals with marked response to gefitinib express a unique pattern of activating mutations in the intracellular domain of EGFR.[31,32] Strikingly, the frequency of these activating mutations is higher in women, non-smokers, Asians, and adenocarcinomas, replicating the clinical observations. This observation suggests once again that the presence of mutations in the target is an indication that the molecule is driving tumor behavior. Further, it suggests that methods to identify patients whose tumors are driven by the target should be developed concurrently with targeted therapeutics if we are to achieve their optimal utility.

    Another agent, erlotinib, has also shown inhibitory activity in multiple tumor cell lines both as a single agent and in combination with chemotherapeutic agents, radiation therapy, or other targeted agents.[33] Initial phase II results for treatment of patients with advanced head and neck cancer (N = 115) showed an objective response in 4.3% in these heavily pretreated patients, regardless of their EGFR status, with a median overall survival duration of 6 months.[34] In this study, as well as in phase II trials in NSCLC and ovarian cancer, the subgroup of patients developing the classic rash showed a significant association with improved survival.[34,35] Phase III trials of erlotinib are underway in NSCLC and pancreatic cancer as a single agent or in combination therapy.[20,21] Preliminary data indicate that, like gefitinib, erlotinib will be most active in patients with specific mutations in EGFR.

    Development of irreversible inhibitors of the erbB family, such as CI-1033 and EKB-569, was pursued to some extent because of the high levels of reversible erbB inhibitors necessary for continuous inhibition of phosphorylation. Covalent bonding of these irreversible inhibitors to the TK domain might sustain inhibition more effectively and for a longer duration at a lower drug level. Additional impetus came from the speculation that reversible inhibitors would not overcome the high intracellular concentrations of ATP. Furthermore, there are as yet unproven expectations that drug resistance emergence might be avoided. Combination therapy with traditional chemotherapeutics and other drug classes also presents an attractive option. However, these agents are in the early stages of investigation, and clinical benefits are unproven.[2,14] Moreover, the long half-life of orally delivered gefitinib or erlotinib suggests that there may be only limited advantages to theirreversible inhibitors. Perhaps 1 reason for the lack of high response rates among these small-molecule EGFR-TK inhibitors is the absence of screening for patients with tumors expressing the EGFR target molecules. Trastuzumab studies did select for patients overexpressing erbB-2 and were able to demonstrate survival benefit.[36] Indeed, had the studies been done in all breast cancer patients without screening for erbB-2 overexpression, the activity of trastuzumab would have been missed due to the confounding effects of the large number of nonresponders. Imatinib also is specific for patients expressing defined kinase abnormalities, such as Bcr-Abl, PDGFR, and Kit. As indicated above, EGFR inhibitors appear to exhibit maximal activity in patients with activating mutations in the intracellular domain. This suggests that selection of patients whose tumors are driven by particular genetic aberrations will be necessary for optimal clinical benefit. Moreover, delay of tumorprogression may be a more appropriate endpoint for clinical trials with these agents, since that was an important finding in preclinical studies.[2] On the other hand, many of these agents are of interest due to their additive and synergistic potential in combination with traditional therapies or other emerging therapies.

  • Figure

    Table 1.

    HER-1/EGFR-TK Small-Molecule Inhibitors

    (Enlarge Slide)

Ras/Raf/MEK/MAPK Pathway

  • Ras proteins occupy a pivotal role in signal transduction, from RTKs and external growth stimuli to a variety of intracellular effector pathways, including the downstream targets of Raf and MEK (a mitogen-activated protein kinase kinase), leading to cell proliferation (see Figure 3). As a response to growth-inducing signals, the inactive membrane-bound Ras-guanosine 5´-diphosphate (Ras-GDP) is switched to the activated membrane-localized Ras-GTP, which subsequently acts on downstream components.[5,37] Normal control of Ras signaling involves GTP hydrolysis by GTPase-activating proteins, which rapidly convert Ras to the inactive Ras-GDP state. To perform signal transduction, the Ras protein also undergoes a series of post-translational modifications that facilitate its localization to the inner surface of the cell membrane and subsequent activity. The first of these modifications requires farnesylation (lipid prenylation) of the Ras protein at a C-terminaltetrapeptide sequence (the CAAX motif) in a step catalyzed by the enzyme farnesyltransferase (FTase). The introduction of this hydrophobic modification allows the Ras protein to more effectively associate with the membrane lipid bilayer. Other protein prenyltransferases, the geranylger-anyltransferases (GGTase) also catalyze prenylation of several proteins, such as Ras.[5,37]

    Mammalian cells have at least 3 ras proto-oncogenes: K-ras, H-ras, and N-ras, which, upon activating mutation, result in constitutive proliferation and anti-apoptotic signaling. Overall, ras mutations have been identified in about 30% of all human cancers, such as the K-ras mutations in NSCLC, colorectal cancer, and pancreatic cancers; H-ras mutations in bladder and kidney cancers; and N-ras mutations in melanomas and hematologic cancers.[38] Ras signaling is also influenced by aberrant upstream signaling, as Ras functions to transmit extracellular signals from growth factors and their receptors to intracellular pathways. With the established involvement of Ras in cancer and the necessity of prenylation for Ras activity, targeting of the FTases with inhibitors presented an attractive strategy to block this pathway.

  •  
  • Several classes of FTase inhibitors exist. Although many have been studied preclinically, few have progressed far in clinical development (Table 2). Early clinical trials of both orally active and parenterally administered FTase inhibitors show similar toxicities, including schedule-dependent myelosuppression and gastrointestinal effects. Studies in solid tumors have also been disappointing. Monotherapy showed no objective responses in tumors, including NSCLC, SCLC, pancreatic, colorectal, and prostate.[1,5] However, preliminary results suggest that R115777 might have some benefit in hematologic malignancies.[42]

    The lack of specificity of the FTase inhibitors suggests that these agents might also inhibit farnesylation of some of the 300 known proteins possessing a CAAX motif. This inhibition might account for some of the antiproliferative activity attributed to these agents.[1,5] Additionally, redundant pathways might subvert the impact of the FTase inhibitors on Ras. Geranylgeranylation of K-Ras and N-Ras after blocking farnesylation may offer a means for continued signal transduction. Thus, although the exact mechanism of action of these agents is not as clear as once thought, preclinical data still suggest that FTase inhibitors might play a role in therapy if properly used.[1,5]

    The downstream MAPK effector pathway, which includes Raf and MEK, also offers sites for drug targeting. B-Raf mutations have been documented in a number of human cancers, including early ovarian cancers and melanomas, and the MAPK pathway is activated in approximately 30% of human malignancies.[44-46] Raf is a family of serine-threonine kinases. Because they can also be activated by protein kinase C alpha and are known to contribute to multidrug resistance expression, they play a large role in oncogenesis.[2] The only known substrates for MEK are the MAPK kinases Erk1 and Erk2 (extracellular signal-regulated kinases), whose substrates are cytosolic and nuclear proteins (probable transcription factors).[5] These downstream kinases have only recently garnered the interest that had previously been reserved for Ras; therefore, inhibitors of these kinases are not as far along in clinical investigations. BAY 43-90006 is an orally available potentinhibitor of Raf-1 that targets the ATP-binding site and is in clinical trials. CI-1040, an inhibitor of MAPK at a non-ATP site, is also in early phase II clinical trials in pancreatic, breast, renal, and colorectal cancers.[1,5] The response rates in renal cell cancers to BAY 43-9006 may, however, represent a rather promiscuous activity of this inhibitor, blocking for example the Kdr VEGF receptor.

  • Figure

    Table 2.

    Farnesyltransferase Inhibitors[5,32]

    (Enlarge Slide)

PI3K/Akt/PTEN Pathway as an Emerging Target

The PI3K pathway plays a role in cell growth; cell cycle progression (both during the G1 phase of the cell cycle and at the G2/M transition); protein translation; sensing the environment, particularly available nutrients and growth factors; motility; neovascularization; metastasis; drug resistance; and cell survival, particularly apoptosis and anoikis (matrix deprivation-induced apoptosis), as well as in transformation induced by many different oncogenes and tumor suppressor genes.[47] Given the importance of these processes in transformation and tumor progression, the PI3K pathway is a potential driver of tumor behavior. Various elements of the PI3K/Akt/PTEN pathway are activated in at least 70% of all cancers, resulting in constitutive activation of the pathway. Indeed, the PI3K pathway is targeted for mutagenesis in human tumors more frequently than any other pathway, with the potential exception of the p53 pathway.[48-50] However, the p53 and thePI3K pathways interact at multiple levels, suggesting that these 2 pathways are really a network and any attempt to distinguish between the pathways is moot. The PI3K pathway is targeted by amplification of the catalytic subunit of PI3K, Akt and S6K1, activating mutations in the catalytic subunit of PI3K and the regulatory subunit of PI3K, inactivating mutations in TSC1/2, LKB1, and PTEN and rearrangements of forkhead and TCL1. In addition, activation of Akt can bypass the effects of radiation and chemotherapy and also of targeted therapeutics such as gefitinib and erlotinib. This has not escaped the attention of the pharmaceutical and biotech industries, where there are multiple programs targeting many of the components of the pathway. CCI-779, RAD001, and AP23573, which target mTOR, a downstream component of the pathway, are in clinical trials and have demonstrated remarkable responses in a subset of patients with glioblastoma multiforme,metastatic melanoma, malignant glioma, mantle cell non-Hodgkin's lymphoma, small cell lung cancer (SCLC), and renal cell carcinoma. Objective response rates have been as high as 10% to 20% in several of the tumors, with stable disease in a significant fraction of patients. Mantle cell lymphoma is particularly intriguing, as response rates may be as high as 44% (see accompanying article by S. Faivre and E. Raymond for a discussion of clinical results with these agents). Why mantle cell lymphoma is highly sensitive is under extensive investigation. Ongoing clinical trials are evaluating mTOR inhibitors in combination with other targeted therapeutics and also in combination with chemotherapy.

Apoptosis Modulation and Antisense Oligodeoxynucleotides

The use of antisense oligodeoxynucleotides (AS-ODNs) involves synthetically producing a short sequence (usually an oligomer of about 20 nucleotides) of chemically modified DNA that will hybridize with a specific mRNA sequence to prevent the translation of the targeted mRNA into protein. It is thought that the mRNA in the AS-ODN complex is subjected to degradation by ribonuclease H, thus removing the mRNA from the pool of translatable RNAs.[51] The AS-ODN enters the cell through endocytosis and, possibly, a receptor-mediated process. The AS-ODN approach has been validated with the approval of the antisense drug fomivirsen for the treatment of cytomegalovirus retinitis in patients with AIDS. A number of AS-ODNs are in clinical development and target proteins, such as H-Ras, c-Raf, and Bcl-2.1 The oncogene Bcl-2 is overexpressed in more than 50% of human cancers.[52] It is known to code for an anti-apoptotic protein that can block cancer cells from undergoingprogrammed cell death (apoptosis) on exposure to anticancer therapies, such as traditional cytotoxic chemotherapy, radiotherapy, and monoclonal antibody therapy. Oblimersen sodium is an 18-mer oligodeoxynucleotide antisense sequence that hybridizes with a key region of Bcl-2 mRNA to block expression.[52,53] Clinical trials of oblimersen in combination with cytotoxic therapies and biologic/immunotherapies are underway in metastatic breast cancer, SCLC, and hematologic malignancies.[30] Initial reports from a phase I study in advanced SCLC showed that oblimersen use in combination with carboplatin and etoposide was well tolerated and produced encouraging results.[54] However, follow-up studies have not been as encouraging.[2] Whether this represents a problem with the approach, failure to deliver stable antisense oligodeoxynucleotides, or selection of a tumor type not dependent on Bcl-2 will require additional investigation.

Small interfering RNAs (siRNA) have demonstrated an ability to downregulate gene expression by targeting RNAs to the RISC complex with subsequent degradation.[55] Although in an earlier stage of evolution, siRNA appears to be a natural gene expression regulator and may have more generalizability than antisense. Small interfering RNAs has shown the ability to regulate gene expression and tumor growth in animal models.

Tumor Angiogenesis Inhibition

Based on evidence that angiogenesis plays a crucial role in tumor growth and metastasis, development of agents that prevent formation of new blood vessels is focused on a number of approaches, of which the following are furthest along: inhibition of VEGF; inhibition of the tyrosine kinase activity associated with the VEGF receptor (VEGFR) on endothelial cells; and inhibition of endothelial cell proliferation. Vascular endothelial cell growth factor is the most frequently upregulated angiogenic growth factor and has been associated with both metastasis and/or poor prognosis in numerous human solid tumors.[56,57] In tumor tissues, VEGF can be constitutively upregulated through both autocrine and paracrine loops with involvement of a wide variety of oncogenes, such as K-ras, p53, WTI, and v-Raf, as well as the growth factors LPA PDGF and IGF-1 and the PI3K pathway described above.[56,57] With recent data supporting the validation ofangiogenesis as an exciting therapeutic opportunity, the National Cancer Institute (NCI) and the Eastern Cooperative Oncology Group (ECOG) have initiated a number of clinical trials for the treatment of melanoma and myeloma, as well as breast, colon, lung, renal, and head and neck cancers, with antiangiogenesis therapies such as bevacizumab and thalidomide as monotherapy and in combination with standard therapies.[58,59]

Bevacizumab (rhuMAB-VEGF) is a recombinant humanized monoclonal antibody directed to bind and neutralize VEGF, thus preventing VEGF from binding to and activating VEGFR on endothelial cells. A report from a phase II study of bevacizumab monotherapy in patients with metastatic renal cell cancer showed that the treatment significantly prolonged time to progression, but not overall survival compared with placebo (P < .001) in patients receiving high-dose antibody (10 mg/kg every 2 weeks).[60] Also promising was the report that bevacizumab (at low [5 mg/kg] or high [10 mg/kg] doses) in combination with fluorouracil/leucovorin as first-line therapy of metastatic colorectal cancer provided higher response rates, longer median time to disease progression, and longer median survival (control, 13.8 months; low-dose bevacizumab, 21.5 months; high-dose bevacizumab, 16.1 months) compared with fluorouracil/leucovorin alone.[61] Although a phase III study ofbevacizumab combined with capecitabine showed a near doubling of the response rate (19% to 30%), the response was not durable and there was no difference in progression-free survival.[62] Since it is thought that earlier use of bevacizumab might provide better outcomes as seen in the trial in colorectal cancer, a new phase III trial has been initiated to investigate bevacizumab combined with paclitaxel in patients newly diagnosed with metastatic breast cancer.[2] Indeed, recent clinical results showing efficacy of bevacizumab in colorectal cancer have resulted in its approval by the US Food and Drug Administration (FDA), making this the first agent to target the vasculature to achieve approval.

Whereas bevacizumab acts directly on VEGF, other small molecules can modulate VEGF activity through selective inhibition of VEGF membrane RTKs. The compounds currently being investigated are competitive tyrosine kinase inhibitors that block the ATP-binding site, preventing downstream signaling. Although most of these molecules have rather broad receptor specificity, the multiplicity of pro-angiogenic factors implicated in tumor angiogenesis suggests that monotherapy will not be sufficient. As a result, these agents are commonly evaluated in combination with conventional chemotherapy.

Vatalanib (PTK787/ZK222584) inhibits all 3 VEGF receptors, as well as PDGFR-beta, c-Kit, and CSF-1R.[63,64] Encouraging results from phase I/II trials in metastatic colorectal, recurrent glioblastoma multiforme, and metastatic renal cell cancer supported further clinical development of this compound. Several phase II studies in a variety of tumor types are ongoing, as well as 2 phase III trials in metastatic colorectal cancer.[28,65-68]

The multi-kinase inhibitors SU11248 and ZD6474 are also progressing in clinical development. In addition to all 3 VEGF receptors, these small molecules inhibit a wide variety of other membrane protein kinases and have demonstrated anti-angiogenic and antitumor effects in animal models.[69] SU11248 is being evaluated in a number of clinical trials in metastatic beast, renal, and colorectal cancers.[30] The demonstrated objective responses after therapy with SU11248 in patients with metastatic GISTs resistant to imatinib supported evaluating this compound in a phase III trial in this patient population.[70] ZD6474 is being evaluated in several phase II trials in both non-small cell and SCLC.

Another area of investigation is vascular targeting agents (VTAs). Rather than inhibiting the growth of new blood vessels, VTAs target the already formed vasculature of tumors, causing the tumor blood vessels to shut down. The ensuing ischemia can cause extensive tumor cell death.[71] However, a thin rim of tumor cells adjacent to normal tissue often survive. Thus, curative regimens using these agents will most likely also include conventional anticancer therapies.

Compared with normal tissues, vascular architecture in solid tumors is abnormal, chaotic, and inadequate in both structure and function.[72] Although these characteristics often impede the effectiveness of conventional chemotherapeutic agents, they provide a unique target for the developing novel antitumor agents.[73] Examples of tumor vascular endothelial targets include prostate-specific membrane antigen (PSMA), VEGF receptors, fibronectin ED-B domain, alphavbeta3 integrin, E-selectin, vascular cell adhesion molecule-1 (VAM-1), and phosphatidyl serine. A number of biological molecules have been developed that target tumor endothelium directly. These molecules are currently being evaluated in preclinical and, in the case of PSMA, early clinical testing.[72,73]

Small-molecule VTAs are further along in clinical development. Two classes of small molecule VTAs are currently being evaluated. The first is flavone-8-acetic acid (FAA) and its derivatives. The mechanism of action of these molecules appears to be indirect through the induction of various modulators of the immune system, such as nitric oxide, serotonin, and tumor necrosis factor (TNF)-alpha.[74] 5,6- dimethylxanthenone-4 acetic acid (DMXAA) is one of the most promising FAA derivatives. In preclinical studies DMXAA demonstrated antitumor activity in a wide variety of murine tumors and induced extensive tumor necrosis in human tumor xenografts.[75] In a phase I study of 63 patients with advanced cancers, a patient with metastatic cervical carcinoma achieved a partial response, and 22 patients achieved stable disease. In 5 patients, the duration of stable disease exceeded 12 weeks. The antitumor activity of DMXAA was observed at well-tolerateddoses.[76]

The second class of small-molecule VTAs consists of tubulin-binding agents. Colchicine, vincristine, and vinblastine are tubulin-binding molecules that have been developed as anticancer agents. The antitumor effects of these agents were uniformly very close to their maximum tolerated dose (MTD). "Second-generation" tubulin destabilizing agents have been developed that disrupt tumor blood vessels at doses well below their MTD.[73] These newer tubulin-binding agents consist of combretastatins and their analogs. Combretastatin A-4 disodium phosphate (CA4DP), the lead compound in this series, has shown potent antivascular and antitumor effects in a wide variety of preclinical tumor models.[77] Three phase I clinical trials of CA4DP involving a total of 96 patients with advanced cancers have been reported.[78-80] Tumor pain was a unique side effect of combretastatin in these studies. In one study, 1 patient with anaplastic thyroid cancer had a completeresponse.[78] Dose dependent reductions in tumor blood flow have been shown by dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and PET at doses that were reasonably well tolerated by patients.[78,81-82]

Several other tubulin-binding agents are also in phase I clinical development, including AVE8062A, ZD6126, and ABT-751.[66] The early clinical evidence of these VTAs warrants further evaluation.

Conclusions

The explosion of new therapeutic targets associated with signal transduction pathways has led to the development of rationally designed drugs and the emergence of biologic-based therapies that have provided both encouraging advances in outcomes and dramatic disappointments. However, interest has not waned, and this area of investigation remains in the early stages of discovery in terms of both the basic science of cellular control and appropriate regimen and patient selection for clinical study. Efforts continue to explore opportunities in this area for medical value. The optimal implementation of these drugs will require the concurrent development of targeted therapeutics with molecular markers able to identify tumors driven by particular targets and molecular imaging approaches able to identify patients who are responding early in therapy.

References

  1. Adjei AA, Rowinsky EK. Novel anticancer agents in clinical development. Cancer Biol Ther. 2003;2(4 Suppl 1):S5-S15.
  2. Syed S, Rowinsky E. The new generation of targeted therapies for breast cancer. Oncology (Huntingt). 2003;10:1339-1351.
  3. Lieberman J, Song E, Lee SK, Shankar P. Interfering with disease: opportunities and roadblocks to harnessing RNA interference. Trends Mol Med. 2003;9:397-403.
  4. Kohn EC, Lu Y, Wang H, et al. Molecular therapeutics: promise and challenges. Semin Oncol. 2004;31(suppl 3):39-53.
  5. Downward J. Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer. 2003;3:11-22.
  6. Rotea W Jr, Saad ED. Targeted drugs in oncology: new names, new mechanisms, new paradigm. Am J Health Syst Pharm. 2003;60:1233-1243.
  7. Vlahovic G, Crawford J. Activation of tyrosine kinases in cancer. Oncologist. 2003;8:531-538.
  8. Kantarjian H, Sawyers C, Hochhaus A, et al. Hematologic and cytogenetic responses to imatinib mesylate in chronic myelogenous leukemia. N Engl J Med. 2002;346:645-652.
  9. Demetri GD, von Mehren M, Blanke CD, et al. Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors. N Engl J Med. 2002;347:472-480.
  10. Sirvent N, Maire G, Pedeutour F. Genetics of dermatofibrosarcoma protuberans family of tumors: from ring chromosomes to tyrosine kinase inhibitor treatment. Genes Chromosomes Cancer. 2003;37:1-19.
  11. Heinrich MC, Corless CL, von Mehren M, et al. PDG-FRA and KIT mutations correlate with the clinical responses to imatinib mesylate in patients with advanced gastrointestinal stromal tumors (GIST). Proc Am Soc Clin Oncol. 2003;22:815. Abstract 3274.
  12. Olayioye MA, Neve RM, Lane HA, Hynes NE. The ErbB signaling network: receptor heterodimerization in development and cancer. EMBO J. 2000;19:3159-3167.
  13. Riese DJ 2nd, Stern DF. Specificity within the EGF family/ErbB receptor family signaling network. Bioessays. 1998;20:41-48.
  14. Janmaat ML, Giaccone G. Small-molecule epidermal growth factor receptor tyrosine kinase inhibitors. Oncologist. 2003;8(6):576-86.
  15. Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science. 1987;235:177-182.
  16. Vogel CL, Cobleigh MA, Tripathy D, et al. Efficacy and safety of trastuzumab as a single agent in first-line treatment of HER2 over expressing metastatic breast cancer. J Clin Oncol. 2002;20:719-726.
  17. Perrotte P,Matsumoto T, Inoue K, et al. Anti-epidermal growth factor receptor antibody C225 inhibits angiogenesis in human transitional cell carcinoma growing orthotopically in nude mice. Clin Cancer Res. 1999;5:257-265.
  18. Cunningham D, Humblet Y, Siena S, et al. Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. N Engl J Med. 2004;351:337-345.
  19. Iressa (gefitinib tablets) Product Information.Wilmington, Del: AstraZeneca Pharmaceuticals LP; 2003.
  20. Available at: www.osip.com/osi/clinical.asp?id=39. Accessed June 23, 2004.
  21. Perez-Soler R. The role of erlotinib (Tarceva, OSI 774) in the treatment of non-small cell lung cancer. Clin Cancer Res. 2004;10(12 Pt 2):4238s-4240s.
  22. Allen LF, Eiseman IA, Fry DW, Lenehan PF. CI-1033, an irreversible pan-erbB receptor inhibitor and its potential application for the treatment of breast cancer. Semin Oncol. 2003;30(Suppl):65-78.
  23. Wissner A, Overbeek E, Reich MF, et al. Synthesis and structure-activity relationships of 6,7-disubstituted 4-anilinoquinoline-3-carbonitriles. The design of an orally active, irreversible inhibitor of the tyrosine kinase activity of the epidermal growth factor receptor (EGFR) and the human epidermal growth factor receptor-2 (HER-2). J Med Chem. 2003;46:49-63.
  24. Xia W, Mullin RJ, Keith BR, et al. Anti-tumor activity of GW572016: a dual tyrosine kinase inhibitor blocks EGF activation of EGFR/erbB2 and downstream Erk1/2 and AKT pathways. Oncogene. 2002;21:6255-6263.
  25. Cohen MH, Williams GA, Sridhara R, Chen G, Pazdur R. FDA drug approval summary: gefitinib (ZD1839) (Iressa) tablets. Oncologist. 2003;8:303-306.
  26. Giaccone G, Herbst RS, Manegold C, et al. Gefitinib in combination with gemcitabine and cisplatin in advanced non-small-cell lung cancer: a phase III trial—INTACT 1. J Clin Oncol. 2004;22:777-784.
  27. Herbst RS, Giaccone G, Schiller JH, et al. Gefitinib in combination with paclitaxel and carboplatin in advanced non-small-cell lung cancer: a phase III trial—INTACT 2. J Clin Oncol. 2004;22:785-794.
  28. Wilkinson E. Surprise phase III failure for ZD1839. Lancet Oncol. 2002;3:583.
  29. Albain K, Elledge R, Gradishar WJ, et al. Open-labeled phase II multicenter trial of ZD 1839 (Iressa) in patients with advanced breast cancer. Breast Cancer Res Treat. 2002;76:S33. Abstract 20.
  30. Available at: www.clinicaltrials.gov. Accessed June 23, 2004.
  31. Lynch TJ, Bell DW, Sordella R, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med. 2004;350:2129-2139.
  32. Paez JG, Janne PA, Lee JC, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science. 2004;304:1497-1500.
  33. Hidalgo M. Erlotinib: preclinical investigations. Oncology (Huntingt). 2003;17(Suppl):11-16.
  34. Soulieres D, Senzer NN, Vokes EE, Hidalgo M, Agarwala SS, Siu LL. Multicenter phase II study of erlotinib, an oral epidermal growth factor receptor tyrosine kinase inhibitor, in patients with recurrent or metastatic squamous cell cancer of the head and neck. J Clin Oncol. 2004;22:77-85.
  35. Clark GM, Pérez-Soler R, Siu L, et al. Rash severity is predictive of increased survival with erlotinib HCl. Proc Am Soc Clin Oncol. 2003;22:196. Abstract 786.
  36. Slamon DJ, Leyland-Jones B, Shak S, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001;344:783-792.
  37. Bishop WR, Kirschmeier P, Braun C. Farnesyl transferase inhibitors: Mechanism of action, translational studies and clinical evaluation. Cancer Biol Ther. 2003(Suppl 1):S96-S104.
  38. Adjei AA. Blocking oncogenic Ras signaling for cancer therapy. J Natl Cancer Inst. 2001;93:1062-1074.
  39. Britten CD, Rowinsky EK, Soignet S, et al. A phase I and pharmacological study of the farnesyl protein transferase inhibitor L-778, 123 in patients with solid malignancies. Clin Cancer Res. 2001;7:3894-3903.
  40. Van Cutsem E, van de Velde H, Karasek P, et al. Phase III trial of gemcitabine plus tipifarnib compared with gemcitabine plus placebo in advanced pancreatic cancer. J Clin Oncol. 2004;22:1430-1438.
  41. Whitehead RP, McCoy S, MacDonald SJ, et al. Phase II trial of R115777 (NSC #70818) in patients with advanced colorectal cancer: A Southwest Oncology Group study. Proc Am Soc Clin Oncol. 2003;22:272. Abstract 1092.
  42. Cortes J, Albitar M, Thomas D, et al. Efficacy of the farnesyl transferase inhibitor R115777 in chronic myeloid leukemia and other hematologic malignancies. Blood. 2003;101:1692-1697.
  43. Dy GK, Bruzek LM, Croghan GA, et al. A phase I trial of the farnesyltransferase (FT) inhibitor, BMS-214662 (B) in combination with paclitaxel (P) and carboplatin (C) in patients with advanced cancers. Proc Am Soc Clin Oncol. 2004;23. Abstract 3066.
  44. Midgley RS, Kerr DJ. Ras as a target in cancer therapy. Crit Rev Oncol Hematol. 2002;44:109-120.
  45. Singer G, Oldt R 3rd, Cohen Y, et al. Mutations in BRAF and KRAS characterize the development of low-grade ovarian serous carcinoma. J Natl Cancer Inst. 2003;95:484-486.
  46. Davies H, Bignell GR, Cox C, et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417:949-954.
  47. Mills GB, Lu Y, Kohn EC. Linking molecular therapeutics to molecular diagnostics: inhibition of the FRAP/RAFT/TOR component of the PI3K pathway preferentially blocks PTEN mutant cells in vitro and in vivo. Proc Natl Acad Sci USA. 2001;98:10031-10033.
  48. Meric-Bernstam, F., Mills G.B. Mammalian target of rapamycin Seminars in Oncology. 2004. In Press.
  49. Drees BE, Mills GB, Rommel C, Prestwich GD. Therapeutic potential of phosphoinositide 3-kinase inhibitors. Expert Opinion on Therapeutic Patents. 2004;14:703-732.
  50. Kohn EC, Lu Y, Wang H, et al. Molecular therapeutics: promise and challenges. Semin Oncol. 2004;31:39-53.
  51. Crooke ST. Progress in antisense therapeutics. Med Res Rev. 1996;16:319-344.
  52. Nahta R, Esteva FJ. Bcl-2 antisense oligonucleotides: a potential novel strategy for the treatment of breast cancer. Semin Oncol. 2003;30(Suppl):143-9.
  53. Klasa RJ, Gillum AM, Klem RE, Frankel SR. Oblimersen Bcl-2 antisense: facilitating apoptosis in anticancer treatment. Antisense Nucleic Acid Drug Dev. 2002;12:193-213.
  54. Rudin CM, Kozloff M, Hoffman PC, et al. Phase I study of G3139, a bcl-2 antisense oligonucleotide, combined with carboplatin and etoposide in patients with small-cell lung cancer. J Clin Oncol. 2004;22:1110-7.
  55. Martinez J, Patkaniowska A, Urlaub H, Luhrmann R, Tuschl T. Single-stranded antisense siRNAs guide target RNA cleavage in RNAi. Cell. 2002 ;110:563-574.
  56. Rak J, Filmus J, Finkenzeller G, Grugel S, Marme D, Kerbel RS. Oncogenes as inducers of tumor angiogenesis. Cancer Metastasis Rev. 1995;14:263-77.
  57. Fernando NH, Hurwitz HI. Inhibition of vascular endothelial growth factor in the treatment of colorectal cancer. Semin Oncol. 2003;30(Suppl):39-50.
  58. Available at: www.cancer.gov. Accessed August 1, 2004.
  59. Sparano JA, Gray R, Giantonio B, et al. Evaluating antiangiogenesis agents in the clinic: the Eastern Cooperative Oncology Group Portfolio of Clinical Trials. Clin Cancer Res. 2004;10:1206-1211.
  60. Yang JC, Haworth L, Sherry RM, et al. A randomized trial of bevacizumab, an anti-vascular endothelial growth factor antibody, for metastatic renal cancer. N Engl J Med. 2003;349:427-434.
  61. Kabbinavar F, Hurwitz HI, Fehrenbacher L, et al. Phase II, randomized trial comparing bevacizumab plus fluorouracil (FU)/leucovorin (LV) with FU/LV alone in patients with metastatic colorectal cancer. J Clin Oncol.2003;21:60-65.
  62. Miller KD, Rugo HS, Cobleigh MA, et al. Phase III trials of capecitabine (Xeloda) plus bevacizumab (Avastin) versus capecitabine alone in women with metastatic breast cancer previously treated with an anthracycline and a taxane. Breast Cancer Res Treat. 2002;76:S37. Abstract 36.
  63. Bold G, Altmann K-H, Frei J, et al. New anilinophthalazines as potent and orally well absorbed inhibitors of the VEGF receptor tyrosine kinases useful as antagonists of tumor-driven angiogenesis. J Med Chem. 2000:43:2310-2323.
  64. Bold G, Frei P, Manley PW, et al. CGP 84738, NVP-AAC789,NVP-AAD777 and related 1-anilo-(pyridylmethyl) phthalazines as inhibitors of VEGF- and bFGF-induced angiogenesis. Drugs Future. 2002;27:43-55.
  65. Reardon D, Friedman H, Yung, et al. A phase I/II trial of PTK787/ZK222584 (PTK/ZK), a novel, oral angiogenesis inhibitor, in combination with either temozolomide or lomustine for patients with recurrent glioblastoma multiforme (GBM). Proc Am Soc Clin Oncol. 2004;23. Abstract 1513.
  66. Steward WP, Thomas A, Morgan B, et al. Expanded phase I/II study of PTK787/ZK222584 (PTK/ZK, a novel, oral angiogenesis inhibitor, in combination with FOLFOX-4 as first-line treatment for patients with metastatic colorectal cancer. Proc Am Soc Clin Oncol. 2004;23.Abstract 3556.
  67. Schleucher N, Trarbach T, Junker U, et al. Phase I/II study of PTK787/ZK222584 (PTK/ZK), a novel, oral angiogenesis inhibitor in combination with FOLFIRI as first-line treatment for patients with metastatic colorectal cancer. Proc Am Soc Clin Oncol. 2002;23. Abstract 3558.
  68. George D, Michaelson D, Oh WK, et al. Phase I study of PTK787/ZK222584 (PTK/ZK) in metastatic renal cell carcinoma. Proc. Am Soc Clin Oncol. 2003;22. Abstract 1548.
  69. Manley PW, Bold G, Bruggen J, et al. Advances in the structural biology, design and clinical development of VEGF-R kinase inhibitors for the treatment of angiogenesis. Biochim Biophys Acta. 2004;1697:17-27.
  70. Demetri GD, Desai J, Fletcher JA, et al. SU11248, a multi-targeted tyrosine kinase inhibitor, can overcome imatinib (IM) resistance caused by diverse genomic mechanisms in patients (pts) with metastatic gastrointestinal stromal tumor (GIST). Proc Am Soc Clin Oncol. 2004;23. Abstract 3001.
  71. Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases. Nature. 2000;407:249-257.
  72. Pilat MJ, McCormick J, LoRusso PM. Vascular targeting agents. Curr Oncol Rep. 2004;6:103-110.
  73. Thorpe PE, Chaplin DJ, Blakey DC. The first international conference on vascular targeting: meeting overview. Cancer Res. 2003;63:1144-1147.
  74. Siemann DW, Mercer E, Lepler S, Rojiani AM. Vascular targeting agents enhance chemotherapeutic agent activities in solid tumor therapy. Int J Cancer. 2002;99:1-6.
  75. Pedley RB, Boden JA, Boden R, et al. Ablation of colorectal xenografts with combined radioimmunotherapy and tumor blood flow-modifying agents. Cancer Res. 1996;56:3293-3300.
  76. Jameson MB, Thompson PI, Baguley BC, et al. Clinical aspects of a phase I trial of 5,6-dimethylxanthenone-4-acetic acid (DMXAA), a novel antivascular agent. Br J Cancer. 2003;88:1844-1850.
  77. Holwell SE, Cooper PA, Thompson MJ, et al. Anti-tumor and anti-vascular effects of the novel tubulin-binding agent combretastatin A-1 phosphate. Anticancer Res. 2002;22:3933-3940.
  78. Dowlati A, Robertson K, Cooney M, et al. A phase I pharmacokinetic and translational study of the novel vascular targeting agent combretastatin a-4 phosphate on a single-dose intravenous schedule in patients with advanced cancer. Cancer Res. 2002;62:3408-3416.
  79. Rustin GJ, Galbraith SM, Anderson H, et al. Phase I clinical trial of weekly combretastatin A4 phosphate: clinical and pharmacokinetic results. J Clin Oncol. 2003;21:2815-2822.
  80. Stevenson JP, Rosen M, Sun W, et al. Phase I trial of the antivascular agent combretastatin A4 phosphate on a 5-day schedule to patients with cancer: magnetic resonance imaging evidence for altered tumor blood flow. J Clin Oncol. 2003;21:4428-4438.
  81. Galbraith SM, Maxwell RJ, Lodge MA, et al. Combretastatin A4 phosphate has tumor antivascular activity in rat and man as demonstrated by dynamic magnetic resonance imaging. J Clin Oncol. 2003;21:2831-2842.
  82. Anderson HL, Yap JT, Miller MP, et al. Assessment of pharmacodynamic vascular response in a phase I trial of combretastatin A4 phosphate. J Clin Oncol. 2003;21:2823-2830.