Cancer Decrease

Cancer Decrease
Successfull treatment

суббота, 20 ноября 2010 г.

Antisense Therapeutics for Tumor Treatment: The TGF-beta2 Inhibitor AP 12009 in Clinical Development Against Malignant Tumors

Overexpression of the cytokine transforming growth factor-beta 2 (TGF-beta2) is a hallmark of various malignant tumors including pancre¬atic carcinoma, malignant glioma, metastasizing melanoma, and metastatic colorectal carcinoma. This is due to the pivotal role of TGF-beta2 as it regulates key mechanisms of tumor develop¬ment, namely immunosuppression, metastasis, angiogenesis, and proliferation. The antisense technology is an innovative technique offering a targeted approach for the treatment of differ¬ent highly aggressive tumors and other diseases. Antisense oligonucleotides are being developed to inhibit the production of disease-causing pro¬teins at the molecular level. The immunothera-peutic approach with the phosphorothioate oli-godeoxynucleotide AP 12009 for the treatment of malignant tumors is based on the specific inhibition of TGF-beta2. After providing pre¬clinical proof of concept, the safety and efficacy of AP 12009 were assessed in clinical phase I/II open-label dose-escalation studies in recurrent or refractory high-grade glioma patients. Me¬dian survival time after recurrence exceeded the current literature data for chemotherapy. Cur¬rently, phase I/II study in advanced pancreatic carcinoma, metastatic melanoma, and metastatic colorectal carcinoma and a phase IIb study in recurrent or refractory high-grade glioma are ongoing. The preclinical as well as the clinical results implicate targeted TGF-beta2 suppression as a promising therapeutic approach for malig¬nant tumor therapy.
16.1    Introduction
Pancreatic cancer is the fourth leading cause of cancer-related deaths in men and women in virtu-ally all industrialized countries (Jemal et al. 2005, 2006). Its incidence cuts across all racial and so-cio-economic barriers. Outcome is nearly always fatal with a 1-year survival rate of about 20% and a 5-year survival rate of less than 5% (Cardenes et al. 2006; Jemal et al. 2005, 2006). The majority of patients exhibit a very aggressive adenocarci-noma (85%). These tumor types are less aggres¬sive and are often curable. Pancreatic tumors are difficult to detect at early stages and, due to their nonspecific symptoms, extremely hard to diagnose. Therefore, most patients present with locally advanced or metastatic disease resulting in high mortality and very short life expectancy. This is at least partially due to the observed resis¬tance of pancreatic cancer to chemotherapy and radiation therapy. Currently, complete surgical resection remains the only therapeutic option with a potential for cure. However, only a low proportion of patients (only 15%-20%) are suit¬able candidates for surgical resection (Cardenes et al. 2006; Siech et al. 2001). The median sur¬vival of these patients who undergo successful resection is approximately 12-19 months with a 5-year survival rate of 15%-20%. Risk factors for pancreatic cancer include advanced age, obesity, diabetes, and chronic pancreatitis (Li et al. 2004; Lowenfels and Maisonneuve 2006). However, cigarette smoking is considered the most signifi¬cant and avoidable risk factor, causing more than 25% of the pancreatic cancer cases (Li et al. 2004;
Lowenfels and Maisonneuve 2006). Because of the lethality of this disease and the failure of standard treatment to date, future efforts will be focused on the advances that are being made in the understanding and delineation of the genetic and molecular cell biology of cancer cells.
During the last three decades, the interest in new therapeutics such as those based on anti-sense technology has strongly increased. Today, the improved technology and ability of chemi¬cal synthesis of antisense oligodeoxynucleotides (ODNs) has become a routine process and offers researchers the possibility to target almost any single gene (Schlingensiepen et al. 1993). This technology has become a powerful research tool in molecular biology, biochemistry, and microbi¬ology, and has tremendous potential in the fields of functional genomics, drug discovery, and clin¬ical therapy, especially oncology. A major cause of cancer lies in defective gene regulation. Such mutations can result either in an overproduction or in abnormal production of proteins promoting dysfunctional growth and tumor development. Antisense drugs are able to block the blueprint (messenger ribonucleic acid, mRNA) of a can¬cer gene and specifically inhibit its conversion into the pathogenic cancer protein. Preventing the formation of such pathogenic factors means combating cancer disease directly at its roots.
Transforming growth factor beta (TGF-beta) is a multifunctional cytokine that has been iden-tified as a key factor in tumor development. TGF-beta is a vital factor controlling several signaling cascades with oncogenic potential, including immunosuppression, epithelial to mesenchymal transition (EMT), metastasis and invasion, an-giogenesis, and proliferation. Overexpression of the TGF-beta isoform TGF-beta2 is a hallmark of various malignant tumors, e.g., pancreatic carcinoma, malignant brain tumors, malignant melanoma, and metastatic colorectal carcinoma. Thus, targeting this key factor to suppress sev¬eral cancer mechanisms simultaneously right at their origin offers a very promising therapeutic approach.
The phosphorothioate oligodeoxynucleotide (S-ODN) AP 12009 is used for the treatment of malignant tumors based on the specific inhibi¬tion of TGF-beta2. This chapter describes this an-tisense technology in general and gives an over-view of oligodeoxynucleotide modifications and their delivery to cells. Furthermore, the status of preclinical and clinical trials with AP 12009 for the treatment of pancreatic carcinoma, malig¬nant glioma, malignant melanoma, and meta-static colorectal carcinoma is presented.

16.2    Targeted Therapies
Until recently the traditional therapy for patients with advanced pancreatic cancer was palliative 5-fluorouracil (5-FU)-based chemotherapy (Au-erbach et al. 1997; Cardenes et al. 2006). Novel approved chemotherapeutic agents such as gem-citabine (Gemzar, Eli Lilly, Indianapolis) and ox-aliplatin (Eloxatin, Sanofi-Aventis, Paris), as well as new therapeutic approaches including tyrosine kinase inhibitors, e.g., erlotinib (Tarceva, Genen-tech, South San Francisco; Osi Pharmaceuticals, Melville, NY) plus chemotherapy, have demon¬strated a survival benefit and improved quality of life in patients with advanced disease (Moore et al. 2005). However, the best combinational therapy still results in median survival of less than 1 year. Furthermore, the high risk of severe side effects and possible resistance to chemother-apeutic agents has evoked considerable interest in molecular pathways of tumors and new treat-ment strategies such as targeted therapies.
Conventional chemotherapeutic treatments aim at rapidly dividing cells. However, even highly proliferative healthy cells such as blood cells, cells in the hair follicles, and cells lining the gastrointestinal tract are attacked. Similarly, con-ventional radiation therapy affects some healthy cells surrounding the radiated tumor during treatment. Newer radiation therapy techniques can reduce but not fully eliminate this damage. This treatment-related damage of healthy tissue induces chemotherapy's and radiotherapy's well-known side effects. Targeted therapy acts by interfering with specific molecules needed for carcinogenesis and tumor growth. Monoclonal antibodies are one example for targeted therapy. Targeted cancer therapies can be more effective and may offer the advantage of reduced treat-ment-related side effects and improved outcomes due to their action restricted only to the target. Recent phase II and III trials with molecular targeted therapies in advanced pancreatic cancer include approaches using monoclonal antibodies [e.g., cetuximab, Erbitux (ImClone Systems, New York; an anti-EGFR), trastuzumab, Herceptin (Genentech, South San Francisco; anti-HER-2), bevacizumab, Avastin (F. Hoffmann-La Roche, Basel; anti-VEGF)], small molecules [e.g., gefitinib, Iressa (Astra Zeneca, London; EGFR inhibitor), erlotinib, Tarceva (EGFR inhibitor)], protein inhibitors (e.g., marimastat or BAY 12-9566, both matrix metalloproteinase inhibi-tors), and antisense therapeutics (e.g., GTI-2501, complementary to the subunit R1 of ribonucleo-tide reductase) (Cardenes et al. 2006; Lee et al.
2006).
Herein we report on the antisense therapy using the S-ODN AP 12009 for the treatment of pancreatic carcinoma and other solid tumors overexpressing TGF-beta2.


16.2.1  The Antisense Mechanism
In 1978 Zamecnik and Stephenson published their exciting results on the successful blockade of the replication of the Rous sarcoma virus by adding a synthetic oligodeoxynucleotide directed against a specific sequence of the viral genome (Zamecnik and Stephenson 1978). Only two de¬cades later, in 1998, the first antisense compound named Vitravene (Novartis Ophthalmics Eu¬rope, Basel; fomivirsen sodium), an S-ODN, was approved by the Food and Drug Administration (FDA) for the treatment of cytomegalovirus-in-duced retinitis in patients with acquired immu-nodeficiency syndrome (AIDS) (Roehr 1998). Especially in the field of oncology, a number of antisense compounds have been developed that are currently in clinical trials for the treatment of different types of tumors (Coppelli and Grandis 2005; Dean and Bennett 2003; Lahn et al. 2005; Schlingensiepen et al. 2006).
In contrast to gene therapy, which aims at replacing, removing, or introducing genes to correct a genetic defect or a mutation, antisense drugs neither alter human genes nor have any ef-fect on genetic information. Antisense molecules are a mirror image of the genetic blueprint or sequence that contains the necessary informa¬tion for the production of the targeted protein. By binding to the blueprint (mRNA), antisense-molecules render the contained information il¬legible, thereby inhibiting the protein produc¬tion (Fig. 16.1).
Different antisense mechanisms are under discussion concerning how the translation of the targeted protein is inhibited; these are (1) sterical blockade of the ribosome (Schlingensiepen et al. 1997), which physically prevents the progres¬sion of splicing or translation, and (2) RNase H-induced mRNA cleavage (Akhtar and Agrawal 1997). RNase H is an endoribonuclease that spe-cifically hydrolyzes the phosphodiester bonds of
the target RNA.


16.2.1.1 Chemical Modifications
of Antisense Oligodeoxynucleotides
Native ODNs contain phosphodiester linkages in their nucleotide backbone making them highly soluble in aqueous solutions but also very sus-ceptible to degradation by exo- and endonucle-ases within minutes (Shaw et al. 1991; Wick-strom 1986). Established modifications of the ODN chemistry aim at an optimal combination of long half-life due to nuclease resistance, suffi¬cient cellular uptake, good hybridization charac¬teristics, specific binding affinity, and reduction of nonspecific interactions, which could cause toxicities (for review see Mahato 2005). The first chemically synthesized modified ODNs were methylphosphonates (Me-ODNs) with a neutral methyl group replacing the negative charge-bear¬ing oxygen of the phosphodiester bond (Miller et al. 1981). Although Me-ODNs demonstrate an excellent nuclease resistance in biological sys¬tems (Tidd and Warenius 1989), their lipophilic nature leads to solubility problems in comparison with other analogs (Brysch and Schlingensiepen 1994; Mahato 2005). Furthermore, this type of oligonucleotides exhibits insufficient duplex for-mation presumably caused by steric hindrance by the methyl group, resulting in poor antisense activity that cannot activate RNase H activity (Crooke 1999; Miller et al. 1981).

In S-ODNs, one of the nonbridging oxygens of the phosphate backbone is substituted by a sulfur atom (Eckstein 1983). S-ODNs show analogous characteristics to unmodified ODNs such as their net charge and aqueous solubil¬ity, but exhibit a significantly higher stability in vitro and in vivo (Shaw et al. 1991). Further¬more, S-ODNs show excellent antisense activity. Pharmacokinetic experiments in rats, mice, and monkeys have shown that S-ODNs are cleared from plasma biphasically (Agrawal et al. 1995). As observed in preclinical models as well as in humans, the pharmacokinetics of S-ODNs are largely independent of the sequence; thus, differ-ent S-ODNs have shown a similar pharmacoki-netic profile (Geary et al. 2001). Immediately af¬ter administration, they are rapidly distributed into different tissues and organs. Major sites of accumulation are liver and kidney followed by spleen, bone marrow, and lymph nodes (Agrawal et al. 1995, 1991; Mahato 2005). Excretion from the human body occurs primarily via the urine, with up to 30% being excreted within 24 h and 70% within 10 days after a single intravenous ad¬ministration (Agrawal et al. 1991, 1995). After intravenous administration, S-ODNs are not de¬tectable in the brain since they are not able to pass the blood-brain barrier (BBB) (Agrawal
et al. 1991).
Further chemical modifications in ODNs in-clude 2'-0-methyl and 2'-0-methoxy-ethyl oli-gonucleotides showing increased nuclease resis¬tance and oligonucleotide:RNA binding affinities (Agrawal et al. 1997). Other chemical modifica¬tions of ODNs such as N3'-P5' phosphoroami-dates and morpholino oligonucleotides enhance stability, target affinity, and bioavailability (Kur-reck 2003). Another class of oligonucleotide-based compounds consisting of small interfer¬ing RNAs has recently become widely used for gene knockdown in vitro and in vivo (Coppelli and Grandis 2005). So far, none of these com¬pounds is in advanced clinical trials. The key fac¬tors are cellular uptake, the therapeutic activity of the individual antisense compounds, and the sequences themselves, rather than the chemical modifications alone.
Apart from selecting the optimal gene area, it is crucial to avoid interaction with proteins via certain base sequences, which may result in non-specific effects. Antisense compounds may con-tain special motifs such as G-quartets or CG-rich sequences (CpG motif). Four consecutive gua-nosines exhibit a nonspecific antiproliferative action and inhibit enzymatic activities in several cell types (Burgess et al. 1995; Yaswen et al. 1993). CpG motifs may activate defense mechanisms in humans, leading to a natural and acquired im¬mune response (Krieg 2002).
The future therapeutic success of antisense compounds will depend, as is the case with any targeted therapy, on the careful selection of op-timal targets, dosing, schedules, and clinical trial design. The ideal drug candidate should drive tumor progression and should not have redun-dant pathways, as is case with PKC, for example.

16.2.2 Delivery of Oligodeoxynucleotides into Cells
Antisense ODNs must be internalized into tar¬get cells in sufficient amounts to exert their in¬hibiting effects by targeted downregulation of RNA encoding disease-inducing genes. Owing to their anionic nature and their size, phosphodi-ester and phosphorothioate ODNs (S-ODNs) are unable to cross the lipophilic cell membrane by passive diffusion. It is well accepted that cellular uptake of S-ODNs is energy-, temperature-, and time-dependent (Levin 1999). The mechanism of cellular uptake can vary depending on the chemical structure and the concentration of the oligonucleotide. Whereas at low concentrations S-ODN uptake is predominantly achieved via a receptor-like mechanism, at higher concentra¬tions adsorptive endocytosis, pinocytosis and ca-veolar potocytosis are described (Lysik and Wu-
Pong 2003; Mahato et al. 2005; Stein et al. 1993;
Zamecnik et al. 1994).
In vitro uptake of free antisense ODNs into cultured cell lines is in some cases inefficient. Depending on the cell type, in vitro uptake of
ODNs is generally enhanced using different vec-tors. A variety of viral and nonviral possibilities of oligonucleotide delivery has been developed for basic and clinical research. Viral vectors in¬clude retroviral, adenoviral, and adeno-associ-ated viral vectors, which introduce their DNA into the cells with high efficiency. A major ob¬stacle of viral vectors in vivo but not in vitro is the host's immune response including both the adaptive response (Yang and Wilson 1995) and the innate immune system (Plank et al. 1996; Sung et al. 2001). Despite the observed limita¬tions on the usage of viral vectors, especially re¬garding safety, they are still the most used gene transfer vehicles (Gardlik et al. 2005).
Nonviral methods make use of cationic lipid complexes, liposomes (see below), polymers, and other reagents. Furthermore, ODNs may be in-ternalized mechanically, i.e., by generating tran-sient permeabilization of the plasma membrane to allow penetration of naked ODNs into cells by diffusion. However, these methods are not useful in vivo and their relevance for gene func¬tion analysis remains questionable. Therefore, plasmid or liposomal complexes are the most commonly used nonviral vectors and repre¬sent attractive tools in gene therapy due to their relatively simple production, low toxicity, and low host immunogenicity (Gardlik et al. 2005). Shen and colleagues demonstrated that the use of a cationic liposome elicits enhanced efficacy of ODNs for the inhibition of TGF-beta2 expres¬sion in the human promonocytic leukemia cell line U937 (Shen et al. 1999). All of these cationic delivery systems internalize ODNs via an endo-cytotic mechanism. In contrast to the in vitro situation, many reports have shown that in vivo uptake of S-ODNs does not depend on cationic carrier liposomes (Braasch and Corey 2002; Tari and Lopez-Berestein 2001).
In vitro, most of the ODNs designated for clinical studies were delivered in the presence of carrier liposomes in order to facilitate the ODN uptake. In contrast, during the selection process toward the development of the herein described antisense S-ODN AP 12009, inhibition of TGF-beta2 expression without carriers was crucial. Im¬portantly, in preclinical experiments performed with and without the carrier protein Lipofectin
(Invitrogen, Carlsbad, CA; a transfection reagent) AP 12009 showed similar effects (see below).

16.3    The Target: Transforming Growth Factor-Beta 2
TGF-beta is a multifunctional cytokine playing various roles in cell functions, including mor-phogenesis, cell proliferation, and migration, and is a key regulator of the immune system. Three isoforms of TGF-beta are described in mam¬mals: TGF-beta1, TGF-beta2, and TGF-beta3. A unique gene on different chromosomes encodes each isoform. All three human isoforms show a different temporal and spatial expression. Major activities of TGF-betas include inhibition of cell proliferation by blocking the cell cycle in late G1 phase, immunosuppressive effects, and enhanc-ing the formation of extracellular matrix. The transcriptional regulation of TGF-beta1 is dif-ferent from that of TGF-beta2 and TGF-beta3 as the latter are mostly under hormonal and devel-opmental control (Roberts 1998).
TGF-beta is synthesized as homomeric pro-proteins in vivo, which need to be activated to bind to the signaling receptors (Murphy-Ullrich and Poczatek 2000; Wakefield and Roberts 2002). The so-called latency-associated protein (LAP) is generated by removal of the N-terminus of the mature TGF-beta by a furin-like peptidase. The LAP is noncovalently associated with a homodi-mer of mature TGF-beta (Li et al. 2006). TGF-beta is secreted as a complex, which consists of the inactive, mature TGF-beta, the LAP, and the latent TGF-beta binding protein (LTBP) (Annes
et al. 2003; Oeklue and Hesketh 2000). Extracel-lular activation of this complex is a critical step in the regulation of TGF-beta function, includ¬ing plasmin-dependent and plasmin-indepen-dent pathways (Derynck and Zhang 2003; Piek et al. 1999; Wakefield and Roberts 2002; Yingling
et al. 2004).
Although TGF-beta1 and TGF-beta2 share various similar receptor binding and signaling properties, some crucial differences have been described. In general, the TGF-beta ligand binds to receptors on the cell surface forming a bi-di-meric receptor complex consisting of two pairs of subunits known as receptor type I (TBR-I, usually ALK5) and type II (TBR-II). A mem¬brane-anchored proteoglycan, known as type III receptor (TBR-III or betaglycan), aids this pro-cess by capturing TGF-beta for presentation to the signaling receptors I and II. Importantly, the type III receptor is particularly important for TGF-beta2, which cannot bind TBR-II indepen-dently and thus depends on the presence of TBR-III to signal—a unique feature that distinguishes TGF-beta2 from TGF-beta1 and TGF-beta3
(Blobe et al. 2001).
The biological activities of TGF-betas are mod-ulated by binding proteins with alpha-2-macro-globulin (A2M) as the major binding protein for TGF-beta1 and TGF-beta2 in plasma (Daniel-pour and Sporn 1990; O'Connor-McCourt and Wakefield 1987). A2M is a homotetrameric gly-coprotein that inhibits various proteinases and serves as a regulator and major carrier of various cytokines (Crookston et al. 1994). It is one of the most abundant proteins in human plasma with a concentration of 2-4 mg/ml. Both isoforms, TGF-beta1 and TGF-beta2, bind reversibly and covalently to native A2M and A2M-methylamine. It has been shown that A2M significantly inhib¬its the receptor binding and biological activity of TGF-betas (O'Connor-McCourt and Wakefield 1987). However, TGF-beta2 is more affected due to a distinctive interaction pattern with A2M compared to TGF-beta1 and other cytokines (Crookston et al. 1994). First, TGF-beta2 reveals substantially higher affinity to A2M and therefore an increased complex formation (Danielpour and Sporn 1990; Liu et al. 2001). Second, it is the only growth factor that binds with equiva¬lent affinity to both A2M and A2M-methylamine (Crookston et al. 1994). In experiments using na¬tive A2M as well as the activated form A2M-me-thylamine, TGF-beta2 shows the highest affinity to both proteins compared to other cytokines including TGF-beta1, nerve growth factor-beta (NGF-beta), platelet-derived growth factor-BB (PDGF-BB), tumor necrosis factor (TNF-alpha), and basic fibroblast growth factor (Crookston et
al. 1994).
The significance of TGF-beta has become in-creasingly evident since it obviously elicits two opposed mechanisms depending on the respec-tive environment (Akhurst and Derynck 2001; Wakefield and Roberts 2002). In normal cells of epithelial origin as well as in early well-dif-ferentiated tumor cells of epithelial origin, the TGF-beta pathway restricts cell growth, differen-tiation, and cell death. However, during progres-sion of cells toward fully malignant tumor cells, these cells undergo changes resulting in reduced expression of TGF-beta receptors, increased ex-pression of TGF-beta ligands, and resistance to growth inhibition by TGF-beta (Moustakas et al. 2002; Wakefield and Roberts 2002).
The crucial role of TGF-beta2 in pancreatic cancer progression and aggressiveness was dem-onstrated in an animal model consisting of human pancreatic cancer cells grown either ectopically in subcutaneous tissue or orthotopically in the pancreas (Choudhury et al. 2004). In this model, TGF-beta2 expression clearly correlated with tu¬mor aggressiveness and metastatic behavior. The far more aggressive orthotopic tumors not only demonstrated a larger size, shorter latent period, higher metastasis, and more extensive invasion of the stomach, but also a higher expression of TGF-beta2 compared to the less aggressive sub¬cutaneous tumors. In another study on human pancreatic tissue samples, immunohistochemi-cal analysis has shown that all three mammalian isoforms of TGF-beta (TGF-beta1, -beta2, and -beta3) were overexpressed (Friess et al. 1993). However, only the TGF-beta2 isoform was sig¬nificantly correlated with advanced tumor stage and a more aggressive phenotype. Pancreatic cancer patients bearing TGF-beta2 producing tumors showed the shortest postoperative sur¬vival period in contrast to patients with tumors producing TGF-beta1, TGF-beta3, or none of the TGF-beta isoforms (Friess et al. 1993).


16.3.1  Targeted Therapy with
the TGF-Beta2 Inhibitor AP 12009

16.3.1.1 Preclinical Experiments
In vitro experiments were performed to evalu¬ate the specificity and efficacy of the TGF-beta2 specific phosphorothioate ODN AP 12009 by employing human tumor cell cultures as well as peripheral blood mononuclear cells (PBMC) from healthy donors and from patients (Schlin-gensiepen et al. 2006).
The efficacy of AP 12009 in reducing TGF-beta2 secretion of human pancreatic carcinoma cells was determined by measuring the TGF-beta2 concentration in culture supernatants us¬ing an enzyme-linked immunosorbent assay (ELISA). Treatment with AP 12009 complexed with the liposomal carrier Lipofectin significantly inhibited TGF-beta2 production compared to Lipofectin alone in all human pancreatic cancer cell lines tested. Importantly, comparable data were obtained in experiments without Lipofectin indicating that AP 12009 alone is able to inhibit TGF-beta induced tumor-promoting effects.
Furthermore, AP 12009 was shown to revert the strong immunosuppressive effects exerted by TGF-beta2. TGF-beta has multiple immunosup-pressive properties including inhibition of T cell proliferation and inhibition of T cell differen¬tiation into cytotoxic T lymphocytes (CTLs) and
helper T cells (Gorelik and Flavell 2001). TGF-
beta inhibits these immune cell functions includ-ing cell-dependent cytotoxicity (Weller and Fon-tana 1995). Treatment with AP 12009 enhances the cytotoxic antitumor response of human lymphokine activated killer (LAK) cells directed against pancreatic carcinoma cells.
The invasion of neoplastic cells into healthy tissue is a pathologic hallmark of highly aggres-sive tumors such as pancreatic carcinoma, malig-nant melanoma, or malignant glioma.
The key mechanism for infiltration of tumor cells into healthy tissue leading to metastasis is tumor cell motility. TGF-beta, produced by tumor cells, acts directly on the tumor cells by in-ducing EMT (Janji et al. 1999), and by increasing motility, invasiveness, and metastasis (Dumont and Arteaga 2000; Oft et al. 1998). AP 12009 in-hibits the migration of cancer cells in vitro. The motility of pancreatic cancer cells was measured employing an in vitro spheroid migration model (Nygaard et al. 1998). Tumor cells spontane¬ously form round shaped clusters (spheroids) when cultured in medium on agar-coated plates, which prevents their adherence to the plastic surface. The spheroids can be transferred into culture medium without agar where the tumor cells start migrating off the spheroids. AP 12009 inhibits migration of the pancreatic tumor cells with the spheroids remaining compact after 24 h. In contrast, untreated and recombinant human
(rh-) TGF-beta2 treated cells migrate and, as a consequence, the spheroids dissolve.
Similar results as described for pancreatic can-cer cells were obtained for other cancer cells in-cluding human malignant glioma and malignant melanoma cell cultures (Jachimczak et al. 1993, 1996; Schlingensiepen et al. 2006). Importantly, all experiments were performed in the presence as well as in the absence of a liposome carrier and showed comparable efficiency to naked and Lipofectin-complexed AP 12009 in various cell lines test.


16.3.1.2 Toxicological Studies
In the current clinical trials of AP 12009 are be¬ing developed for the treatment of TGF-beta2-overproducing tumors such as malignant gli-oma, pancreatic carcinoma, metastatic colorectal carcinoma, and metastatic melanoma. Whereas AP 12009 is administered systemically by intra¬venous infusion in the indications for pancreatic carcinoma, metastatic colorectal carcinoma, and melanoma, in the case of high-grade glioma the same substance is applied locally by convection-enhanced delivery (CED) directly into the brain tumor tissue.
Local toxicity studies applying AP 12009 by the intrathecal and intracerebral routes were performed in rabbits and monkeys in order to match the intended human mode of applica¬tion in malignant glioma as close as possible. AP 12009 showed excellent local tolerability in rabbits and monkeys when administered by intra-thecal bolus injection. Neither clinical signs of toxicity nor substance-related histomorphologi-cal changes were observed. The application of AP 12009 via continuous intracerebral infusion focally resulted in a mild to moderate lympho-cytic leptomeningo-encephalitis. Changes are considered a reversible immunological reaction to AP 12009. Local tolerance tests of AP 12009 in rabbits after intravenous, intraarterial, intramus-cular, paravenous, and subcutaneous application led neither to macroscopic nor to microscopic changes.
Acute toxicology studies in mice and rats as well as subchronic toxicity studies in rats and in cynomolgus monkeys were performed employ¬ing intravenous infusion. Liver and kidney were identified as target organs. The observed changes match the common toxic effects reported for S-ODNs (Henry et al. 1997; Levin et al. 1998). Detailed methods and results were reported by Schlingensiepen et al. (2005).
The pharmacological effects of AP 12009 on the cardiovascular system, complement ac-tivation, and hematological parameters corre-sponded well to the effects reported for other phosphorothioate ODNs as a class of compounds
(Mahato 2005).
AP 12009 showed neither mutagenic effect in the Salmonella typhimurium strains nor indica¬tions of mutagenic properties in cultured human peripheral lymphocytes with respect to chro-mosomal or chromatid damage. Furthermore, AP 12009 showed no mutagenic properties in the mouse bone marrow micronucleus study us¬ing intravenous administration.


16.3.1.3 Clinical Studies: Systemic Application
In pancreatic carcinoma cells, all three mam-malian isoforms of TGF-beta (TGF-beta1, TGF-beta2, and TGF-beta3) are expressed. However, only excessive expression of TGF-beta2 is signifi¬cantly associated with pancreatic cancer progres¬sion (Friess et al. 1993).
Spurred by the clinical data in recurrent or refractory high-grade glioma patients (see Sect. 3.1.4) and the impressive antitumor ac¬tivity in a wide variety of preclinical assays (Schlingensiepen et al. 2006), the clinical stud¬ies for other solid tumors were initiated. A mul-ticenter dose-escalation phase I/II trial with AP 12009 in adult patients suffering from ad-vanced pancreatic carcinoma (AJCC stage IVA or IVB) as well as metastatic melanoma (AJCC/ UICC stage III or IV) and advanced metastatic colorectal carcinoma (AJCC/UICC stage III or IV), is currently ongoing. The primary endpoint is the assessment of the maximum tolerated dose (MTD) as well as the dose-limiting toxicities. Secondary objectives include safety and tolerabil-ity of AP 12009 and its potential antitumor activ¬ity. Adult patients (18-75 years) with advanced tumors who are not or no longer amenable to established therapies are eligible for this dose¬escalation study. Karnofsky performance status (KPS) should be at least 80%. Patients receive the study drug intravenously via an implanted port system at weekly intervals. Up to ten treatment cycles are to be applied per patient.
The majority of patients already treated re-ceived more than the minimum number of two cycles. One of them received ten full cycles. So far, AP 12009 revealed a good safety profile. The MTD has not yet been reached. Further dose es-calations are ongoing.

16.3.1.4 Clinical Studies: Local Application in High-Grade Glioma Patients
The TGF-beta2 isoform is specifically overex-pressed in malignant gliomas (Frankel et al. 1999; Maxwell et al. 1992). The increased levels of TGF-beta2 are associated with disease stage and causative for the immunodeficient state of patients (Bodmer et al. 1989; Kjellman et al.
2000; Maxwell et al. 1992).
In three phase I/II dose-escalation stud¬ies (G001, G002, and G003) a total of 24 adult patients with recurrent or refractory malig¬nant glioma, i.e., anaplastic astrocytoma (AA,
WHO grade III) or glioblastoma (GBM, WHO
grade IV), and evidence of tumor progression were treated with AP 12009 (Schlingensiepen et al. 2006). In these studies, the drug was ad-ministered intratumorally using CED over a 4- or 7-day period. The CED application allows AP 12009 to bypass the BBB. The BBB serves as a natural defense system by blocking the entry of foreign substances, including bacteria and toxins but also many therapeutic agents (Bobo et al. 1994). While conventional diffusion is characterized by a steep drop in drug concentra-tion close to the catheter tip, CED creates a ho-mogeneous drug concentration extending over several centimeters in diameter (Lieberman et al. 1995). To facilitate multiple cycles of AP 12009, the investigational drug was infused through an implanted port system connected to the intratu-moral catheter. AP 12009 proved to be well toler¬ated and revealed a good safety profile. Since two complete remissions in two different dose groups were observed (see below), further dose escala¬tion was not necessary. MTD was not reached.
Although the clinical phase I/II trials were pri-marily designed to assess safety, survival times as well as tumor response data were obtained. Data on antitumor activity from 24 patients included several patients with stabilization of disease and two patients with complete tumor remission, both of them long-lasting without recurrence. One of these two patients, diagnosed with AA, was treated with only one course of AP 12009. At baseline four tumor lesions had been detected, which were spread over both hemispheres. Only one lesion had been infused with one cycle of AP 12009, but all lesions had disappeared several months after start of treatment despite an ini¬tial and temporary increase in tumor volume at the beginning of the treatment. The patient died from a myocardial infarction without any signs of tumor, 25 months after start of AP 12009 treat¬ment. The second patient, also diagnosed with AA, received a total of 12 cycles of AP 12009 over the course of the three phase I/II studies (G001,
G002, and G003; Fig. 16.2).
Prior to AP 12009 treatment, he had been treated with surgery, radiation, and chemo-therapy [temozolomide (TMZ) after the first re-lapse], followed by a second incomplete surgery. After an initial stabilization following the second cycle, the enhancing lesion continued to increase until 10 months after baseline G001 (Fig. 16.2b), inducing a significant edema. The central read¬ing of the magnetic resonance image (MRI) 20 months after the start of AP 12009 treatment (in G001) was evaluated as partial response (PR, 83% tumor reduction, Fig. 16.2c); there was com-plete response after 22 months. The patient is known to still be alive today; the MRI in August 2006 (Fig. 16.2d) showed no recurrence. Survival of this patient after the first recurrence is now 307 weeks (71 months); it has been 286 weeks (66 months) since treatment with AP 12009 be¬gan (status 01 August 2007).
As of 01 August 2007 the median overall sur-vival after recurrence for AA patients treated with AP 12009 was 146.6 weeks (range 32.0¬306.6 weeks), and for GBM patients treated with AP 12009 44.0 weeks (range 18.9-87.9 weeks). The most recent and accurate survival data after start of therapy that clearly distinguish between recurrent AA and GBM are available for the current gold standard treatment TMZ. The re-

ported median overall survival for TMZ alone is 42.0 weeks (9.7 months) for recurrent AA (The-odosopoulos et al. 2001), and 31.8 (7.3 months) (Yung 2000; Yung et al. 2000) or 32.0 weeks (7.4 months) (Theodosopoulos et al. 2001) for recurrent GBM. These results were reported for patients with high-grade glioma who received TMZ as first treatment after recurrence. In the adjuvant treatment of newly diagnosed glioma, the combination of TMZ with radiotherapy has improved median overall survival from 12.1 to 14.6 months (Stupp et al. 2005).
The phase IIb clinical trial of AP 12009-G004 is an international, open-label, active-controlled dose-finding study in high-grade glioma pa¬tients. The main trial objective is the compari¬son of two different doses of AP 12009 (10 uM or 80 uM) against standard chemotherapy. In all, 145 patients with either recurrent or re¬fractory AA (WHO grade III) or GBM (WHO
grade IV) are receiving either one of the two doses of AP 12009 or standard chemotherapy [TMZ or procarbazine/CCNU (lomustine)/ vincristine = PCV, if TMZ was already given].
AP 12009 is applied intratumorally by CED dur-ing a 6-month active treatment period at weekly intervals. The primary efficacy endpoint is tumor response after radiological evaluation. The main secondary efficacy endpoints are overall survival and 12-month survival. As in the previous stud-ies, preliminary data show long-lasting responses both in recurrent or refractory AA and GBM
patients (Bogdahn et al. 2006; Hau et al. 2006).
Especially in recurrent or refractory AA patients, very promising efficacy data have been docu-mented compared to current standard treatment
with TMZ or PCV.

16.4   Summary
Despite tremendous advances in cancer research and the development of new therapies, patients with malignant tumors such as advanced pancre-atic carcinoma, metastatic melanoma, metastatic colorectal carcinoma, and malignant glioma still face a poor prognosis. The severe morbidity and mortality of these malignant tumor types makes the identification of factors associated with their incidence an important area of both preclinical and clinical research. Antisense technology is a new and innovative method offering a causal approach for the treatment of various highly ag-gressive diseases. Antisense compounds inhibit the production of disease-causing proteins at the molecular level and combat tumor development directly at its roots. Preclinical experiments us¬ing the TGF-beta2 specific phosphorothioate
ODN AP 12009 revealed the potential of this
compound to reverse TGF-beta2 induced immu-nosuppression as well as inhibition of tumor cell proliferation and tumor cell migration. Initial clinical studies have demonstrated AP 12009 to be well tolerated and safe. Furthermore, the first evidence of efficacy of AP 12009 antisense ther¬apy in recurrent or refractory high-grade glioma has been provided.
These data confirm that the blockade of TGF-beta2, a key factor in tumorigenesis, in tumor tissue by AP 12009 represents a novel and promising therapeutic approach for malignant tumors such as advanced pancreatic carcinoma and malignant glioma. This approach aims at a reduction of tumor-promoting effects and, most importantly, an enhancement of the antitumor immune response.
 

Targeted Therapy of the Epidermal Growth Factor Receptor

The epidermal growth factor receptor (EGFR)-mediated pathway is one of the most promising targets for the development of new strategies in anticancer treatments. The so-called "small molecule" tyrosine kinase inhibitor erlotinib has gained marketing authorization in the United States for advanced adenocarcinoma of the lung and for pancreatic cancer, whereas the antibody cetuximab is registered for metastatic colorectal cancer and cancers of the head and neck. Ongoing studies are evaluating the impact of EGFR-targeting therapy in the treatment of locally advanced and metastatic pancreatic cancer.

15.1    Introduction
Epidermal growth factor receptor (EGFR) is known to be of high importance in the development of tumors and their survival in the organism. It belongs to the family of protein tyrosine kinases that have a big influence on the cellular regulation of growth, differentiation, and apoptosis (Ullrich and Schlessinger 1990). Located on the cellular surface, the molecule mediates signal transduction pathways by responding to extracellular signals. Via signal transduction cascades it interacts with different molecules regulating cell proliferation, survival, differentiation and migration (Prenzel et al. 2001; Sonenshein 1997; Alessi and Cohen 1998). Finally, the signal reaches the nucleus, regulating gene expression and transcription (Bromberg and Darnell 2000). Furthermore, EGFR interacts with other members of its receptor family, such as e.g. HER2neu/ErB2 (Gschwind et al. 2001; Earp et al. 1995).
In normal tissue, EGFR is largely controlled by a diversity of regulating mechanisms. In can¬cer tissue, however, these control mechanisms fail, resulting in overexpression and activation (Salomon et al. 1995; Xu et al. 1984). As in other epithelial tumor types, in pancreatic cancer over-expression of EGFR is correlated with bad clini¬cal outcomes due to increased tumoral aggres¬siveness (Yamanaka et al. 1993).
EGFR plays an important role in the develop-ment of pancreatic cancer. In over 90% of cases EGFR is overexpressed (Lemoine et al. 1992), stimulating pancreatic tumor cell growth (Funa-
tomi et al. 1997).
Another important EGFR activity concerns angiogenesis. EGF induces vascular endothelial growth factor and so, in addition to its direct tumor growth induction, it enhances tumor growth by supporting vascularization (Goldman
et al. 1993).
Taking into account all these activities in¬duced by EGFR—regulation of cell growth, tu-morigenesis, and angiogenesis—it is obvious that blocking EGFR activity represents an attractive anticancer treatment approach.
Two ways of EGFR blockade have been devel-oped. One class is represented by small molecule tyrosine kinase (TK) inhibitors, which block adenosine triphosphate binding and so inhibit TK activity. Many of these TK inhibitors have been synthesized, some have been developed clinically, such as gefitinib, lapatinib, and erlo-tinib. For pancreatic cancer, erlotinib is the most developed compound.
The other strategy is inhibiting ligand binding to EGFR by monoclonal antibodies and thereby blocking the following signal transduction cas-cade, including, e.g., cetuximab or matuzumab. As for pancreatic cancer, the most important compound is cetuximab.

15.2    Preclinical Studies
The above-mentioned findings were investigated on a cellular level in several preclinical studies to evaluate EGFR-targeted therapy for human pancreatic cancer. Ng et al. (2002) showed a sig-nificant increase in apoptosis in SCID (severely combined immunodeficient) mice bearing a pan-creatic cancer xenograft. Animals were treated with a combination of gemcitabine, wortmannin (phosphatidylinositol 3'-kinase inhibitor), and erlotinib given intravenously.
In another experiment erlotinib alone showed a significant decrease of proliferation of human pancreatic cancer cells (HPAC) in vitro. Ortho-topically human pancreatic cancer (HPAC)-implanted nude mice showed reduced tumor implantation, size, weight, and jaundice when treated with erlotinib (Durkin et al. 2006).
Additional effects could be shown in experi-ments performed with cetuximab alone or in a combination of cetuximab with gemcitabine. In L3.6pl tumors implanted in the pancreas of nude mice, growth inhibition and tumor regression up to complete tumor disappearance was docu-mented for either cetuximab alone or in com-bination with gemcitabine. No liver metastases were seen in the combination group whereas 50% of the control group (no therapy) showed hepatic spread of the disease. Interestingly, ther¬apy using cetuximab decreased the production of vascular endothelial growth factor and inter-leukin-8 significantly. Consequently, cetuximab reduced microvessel density and increased the percentage of apoptotic endothelial cancer cells. These effects were potentiated when combined with gemcitabine.
Preclinical investigations demonstrated a strong rationale for EGFR-targeting strategies against cancer of the pancreas and were therefore translated into a variety of clinical protocols in combination with chemotherapy or radiotherapy (or both).
15.3    Clinical Trials in Pancreatic Cancer
Standard of care in pancreatic cancer is chemo-therapeutic treatment with gemcitabine. How¬ever, response rates and survival data are poor (Burris et al. 1997) and there is a strong clinical need for improved systemic therapy. Few data are available on compounds interfering with EGFR in pancreatic cancer. These clinical studies will be discussed here.


15.3.1  Clinical Trials with TKIs
15.3.1.1 Erlotinib (Tarceva) in First-Line Treatment of Pancreatic Cancer
Moore et al. (2007) compared gemcitabine plus erlotinib (Tarceva, Roche Pharmaceuticals, Ba¬sel) versus gemcitabine plus placebo in a phase III trial. From 176 sites in 17 countries, 569 patients were randomized to receive either gemcitabine 1,000 mg/m2 weekly on days 1 to 43 (i.e., for 6 weeks), followed by 1 week's rest and then, on days 1, 8, and 15 of a 4-week cycle with erlotinib, given at a dose of 100 or 150 mg/day orally or plus placebo. The primary endpoint of the trial was overall survival; secondary endpoints in-cluded progression-free survival (PFS), response rate, response duration, toxicity, and quality of life. Response was evaluated every 8 weeks using Response Evaluation Criteria In Solid Tumors
(RECIST) criteria.
An interim safety analysis was performed once 50 patients had shown no major increase in toxicity after receiving gemcitabine plus erlotinib at 100 mg/day. Thus, accrual at 150 mg/day was opened. However, recruitment in the 100-mg/ day group was so fast that by the time of the in¬terim analysis for the 150-mg/day group the trial was almost completed with patients on 100 mg/ day. It was therefore decided to include the ap-propriate number of patients for 80% statistical power for the 100-mg/day group.
Survival analysis included 486 and showed a significantly longer overall survival for the gemcitabine plus erlotinib group than for gem-citabine alone with an estimated hazard ratio of 0.82 (95% CI 0.69-0.99, p = 0.038). Median sur¬vival times were 6.24 vs 5.91 months, 1-year sur¬vival rates were 23% (95% CI 18%-28%) and 17%
(95% CI 12%-21%), respectively. Progression-free survival was also longer in the gemcitabine-erlotinib arm (median 3.75 vs 3.55 months, HR 0.77, p = 0.004). Response rates slightly favored gemcitabine-erlotinib (ORR 8.6% vs 8%) with a duration of response of 163 days in both arms.
The treatment was generally well tolerated and comparable in both treatment arms. Known EGFR-induced side effects (Shepherd et al. 2005) included rash, diarrhea, and ILD-like symptoms were more common in the gemcitabine-erlotinib arm, but usually mild to moderate. Hematotoxic-ity did not differ between treatment arms (Grade 3/4 neutropenia 24% vs 27%, thrombocytopenia 10% vs 11%). The toxicity profile of both com-pounds did not differ to that known as single agents.
Quality of life was comparable between both treatment arms.
As seen before with EGFR inhibitors (Xiong and Abbruzzese 2002), the presence of rash in the gemcitabine-erlotinib arm was correlated with a greater likelihood of treatment response.
This significant improvement in overall sur-vival of 22% compared to gemcitabine plus pla-cebo led to marketing authorization in the USA for erlotinib in combination with gemcitabine in
2006.
At the ASCO 2007 Gastrointestinal Cancer Symposium, a phase I of a triple therapy com-bining erlotinib, the monoclonal VEGF receptor antibody bevacizumab, and gemcitabine was re-ported. In 12 patients the combination seemed to be well tolerated, and the maximum tolerated dose (MTD) has not yet been reached. Two pa-tients showed partial response, 4 had stable dis-ease (Gomez-Martin et al. 2007).

15.3.1.2 Erlotinib (Tarceva) in the Treatment of Relapsed Pancreatic Cancer
As patients who fail standard first-line treatment with gemcitabine have no standard treatment options, the impact of new compounds is being examined in this population.
At the ASCO 2007 Gastrointestinal Cancer Symposium, a trial of single-agent erlotinib for patients with relapsed pancreatic cancer has been reported. In 13 patients having received two prior lines of chemotherapy (1-5), erlotinib showed clinical activity in 5 patients, resulting in stabili-zation or improvement for up to 12 months. No grade IV toxicity was described; 4 patients had grade 2 rash, 2 had grade 3 diarrhea (Epelbaum
et al. 2007).
Another trial examined the impact of capecitabine plus erlotinib after failure of a gemcitabine-based chemotherapy in pancreatic cancer. Receiving capecitabine, 2,000 mg/m2, plus erlotinib, 150 mg per day, were 28 patients. The toxicity profile met the expectations with grade 3/4 rash and diarrhea (14% each), hand-foot syndrome (11%), and stomatitis (7%). Par¬tial response was reported in 11% and stable dis¬ease in 57% of patients. The median survival was 6.7 months (Blaszkowsky et al. 2005).

15.3.1.3 TK-Inhibitors in Combination with Radiation Therapy in Pancreatic Cancer
The combination of the TK inhibitor gefitinib, capecitabine, and radiotherapy led to an in-creased toxicity in 10 patients with pancreatic cancer without responses in a phase I dose-finding study (Czito et al. 2006). When given simultaneously to radiation therapy, 825 mg/m2 capecitabine plus 250 mg/day gefitinib induced dose-limiting toxicity in all 3 patients on this dose level (grade 3 nausea, diarrhea), resulting finally in patient withdrawal. On dose level 1 (capecitabine 650 mg/nr* + gefitinib 250 mg/day) DLTs appeared in 3/7 patients. As for efficacy, no responses have been reported; stable disease was reported for 6 out of 7 patients on dose level 1, and 1 out of 3 patients on dose level 2. No patient was converted to resectable status.

15.3.2 Clinical Trials with Anti-EGFR Antibodies
15.3.2.1 Cetuximab (Erbitux) in the First-Line Treatment of Pancreatic Cancer
Cetuximab (Erbitux, Merck, Darmstadt, Ger-many) has also been combined with standard gemcitabine chemotherapy as a first-line treat-ment of pancreatic cancer. A phase II trial has been performed in 41 patients, receiving cetux-imab (initial dose 400 mg/m2, then 250 mg/m2 weekly) followed by gemcitabine 1,000 mg/m2 weekly for 7 weeks plus 1 week's rest. Subsequent cycles were 4 weeks long with gemcitabine given on days 1, 8 and 15 and a rest on day 22. Patients were treated until progression or intolerable tox-icity.
Patients had a median Karnofski score of 80%; 85.4% had metastatic disease.
Out of 41 patients, 5 (12.2%) showed partial response, and a further 26 (63.4%) had stable dis-ease, resulting in a disease control rate of 75.6%.
As discussed above, the effect of EGFR block-ade combines the reduction of tumor growth with antiangiogenesis, both of which in the short run do not result in tumor shrinkage but appear clinically as disease stabilization. Furthermore, the accurate assessment of pancreatic tumor imaging is difficult due to its anatomic location. Therefore, time to progression, which includes objective response plus stable disease, and overall survival may be more appropriate parameters for tumor response assessment in pancreatic cancer.
Median time to best response was 1.7 months, median duration of response was 3.8 months, duration of disease control was 5.4 months, and median time to progression 3.8 months. Median survival duration was 7.1 months; the 1-year overall survival rate was 31.7% and the 1-year PFS rate 12% (Xiong et al. 2004).
These data clearly exceed the results of gem-citabine monotherapy, which showed a 1-year survival rate of 18% and a 1-year PFS of 9%. Also, median TTP and overall survival (3.8 and 7.1 months, respectively) favor the combina-tion of cetuximab and gemcitabine (Burris et al.
1997).
Response data, PFS, and overall survival of the combination cetuximab plus gemcitabine are also comparable to those of other EGF-targeted therapies combined with gemcitabine, as men-tioned above.
Acne-like rash (85.4%), asthenia (85.4%),
nausea (61%), weight loss (58.5%), diarrhea (53.7%), abdominal pain (53.7%), and vomiting (51.2%) were the most common adverse events. As for grades 3 and 4, neutropenia (39%), asthe-nia (22%), abdominal pain (22%), and throm-bocytopenia (17.1%) were the most commonly reported severe toxicities.
One trial explored the efficacy and safety of cetuximab (250 mg/m2 per week) combined with gemcitabine (1,000 mg/m2 q2w) and oxalipla-tin (100 mg/nr* q2w) in 43 patients as first-line treatment of metastatic pancreatic cancer (GE-MOXCET). Toxicity was mainly hematological with grades 3/4 of leucopenia 10%, anemia 15%, and thrombocytopenia 12%. Nonhematological toxicity involved nausea 17%, infection 16%, di¬arrhea 7%, and allergy 6%. Of the patients, 71% had skin rash, 5% grade 3. The overall response rate (34 evaluable patients) was 38% with 1 com¬plete and 12 partial responses. In all, 9 patients
(26%) had stable disease. GEMOXCET has been
shown to be a feasible regimen with moderate toxicity and promising efficacy data (Kullmann
et al. 2007).

15.3.2.2 Cetuximab (Erbitux) in Combination with Radiation Therapy in Pancreatic Cancer
For locally advanced pancreatic cancer, chemo-radiation using gemcitabine or 5-fluouracil plus radiotherapy, is standard care, but results are poor with overall survival of between 7 and 12 months and 1-year survival rates of 30%-45% (Tsai et al. 2003). Given the promising results of EGFR targeting therapy in pancreatic cancer, the introduction of cetuximab in this setting as a novel treatment strategy was a stringent thera¬peutic demand.
Two phase II trials examined the combination of gemcitabine, cetuximab, and radiotherapy as a preoperative induction treatment (Pipas
et al. 2006; Krempien et al. 2006) (PARC trial
ISRCTN56652283). Though still small in num¬ber and preliminary (10 and 20 patients, respec-tively), the first data report shows promising re-sults, with tumor control rates of 80% and 85%, rendering 60% of patients operable by tumor downstaging. The toxicity profile did not differ from that known for gemcitabine and cetuximab, with rash, diarrhea, and hematotoxicity as the main adverse events. Therefore, even as a neoad-juvant approach, the combination of gemcitabine plus cetuximab is feasible and warrants efficacy. The PARC trial will include 66 patients with lo¬cally advanced nonresectable pancreatic cancer, evaluating feasibility via the toxicity profile as its primary study aim, and response rates, time to progression, downstaging of tumor size, post-therapy resectability, and quality of life will be its secondary study aims.

15.4    Conclusion
EGFR-targeting therapies with TK inhibitors and monoclonal antibodies such as cetuximab repre-sent a substantial improvement in the treatment options for patients with pancreatic cancer and probably will enhance the chances and the prog-nosis of those patients.
 

Antiangiogenic Strategies in Pancreatic Cancer

Since the introduction of gemcitabine in the treatment of pancreatic cancer, progress in the use of combination chemotherapies has been very limited. Of the different novel options, anti-angiogenic treatment strategies are among those being intensively studied in preclinical and clini¬cal settings of adenocarcinoma of the pancreas. Phase I and limited-size phase II studies using drugs with antiangiogenic properties have re¬ported encouraging results. Overall, the results of phase III studies with some metalloprotease inhibitors and bevacizumab have so far failed to demonstrate a survival benefit for these drugs. Further investigations that will take into account the heterogeneity of pancreatic cancer are war¬ranted using these or other antiangiogenic active substances.

14.1    Introduction
Targeted therapies inhibiting specific pathways that facilitate the growth and progress of malig-nant tumors are under intensive investigation in oncology. So far, several agents developed to in-hibit tumor angiogenesis have been evaluated in clinical studies [11, 20]. In this context advanced adenocarcinoma of the pancreas represents a malignancy worth studying.
The highly aggressive nature of pancreatic cancer may be due to the expression of growth factors, resulting in high intrinsic tyrosine kinase activities that stimulate cell proliferation, dis-semination, and neoangiogenesis. A number of studies have indicated that angiogenesis involves several cytokines such as vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), and angiopoietin 1 [1, 4, 21, 28]. VEGF and bFGF are clearly overexpressed in pancreatic cancer and result in accelerated tumor growth [29, 46]. The cellular effects of these fac-tors are mediated by specific cell surface recep-tors with intrinsic tyrosine kinase activities.
The results of qualitative and semiquantita-tive assessments of VEGF and VEGF receptor expressions in pancreatic cancer have been con¬firmed by quantitative methods, demonstrating that VEGF and its two principal receptors were expressed in adenocarcinomas of the pancreas [8, 9]. These expressions vary in different indi¬vidual tumors, but a significant association was found between expression of VEGF-R2 and poor prognosis, suggesting that VEGF-R2 expression is a marker of more aggressive disease [8]. Both VEGF and VEGF-receptors are overexpressed in pancreatic cancer, and the degree of expression correlates with microvessel density and poor prognosis [40]. This is true not only in advanced metastatic pancreatic adenocarcinoma but also for early recurrences after curatively intended resection [37].
In preclinical models, inhibition of VEGF decreased pancreatic cancer growth by inhibi¬tion of neovascularization and lymphangio-genesis [4, 9, 31]. In addition, VEGF inhibition decreases interstitial pressure within the tumor and thereby increases the delivery of chemo-therapeutic agents, suggesting an additive effect of antiangiogenic and conventional cytotoxic therapies [4, 50].
In addition to synergistic interactions with other treatment modalities, inhibition of tumor angiogenesis theoretically offers several benefits such as a lack of severe toxicities familiar to cy-totoxic therapies, a lack of tumor resistance, and thus a potent antitumor effect. It has been dem¬onstrated that a disruption of VEGF signaling, for example by the use of neutralizing antibod¬ies, slows tumor growth in preclinical models. It is important to realize that the antiangiogenetic suppression of VEGF signaling targets the nutri¬tional support of tumor cells by inhibiting blood vessel formation.
Treatment with inhibitors of angiogenesis most likely will prevent tumor progression, fa¬cilitate tumor cell killing by cytotoxic agents, radiation, or immunological strategies, and may induce tumor cell senescence and stimulate apoptosis. In addition to the antitumor efficacy of antiangiogenic agents demonstrated in pa¬tients with different tumor types and the antitu¬mor effect of cytotoxic drugs, the combination may result in a supra-additive effect.
While an increasing number of new drugs for antiangiogenetic strategies have been developed, it has become clear that the classical cytotoxic drug that is effective in the treatment of patients with pancreatic cancer, gemcitabine, may have antiangiogenetic effects too.

14.2    Results in Preclinical Models or Phase I Studies in Patients
Small molecule tyrosine kinase inhibitors such as PD173074 block VEGF-R2 as well as bFGF-R1 signaling, thus showing antiangiogenetic and antimitogenic activities and abrogating two im-portant pathways in cancer growth and metas-tasis [9]. In human pancreatic cancer cell lines, PD173074 inhibited cell growth in correlation with the level of bFGF-R1 expression of the tu¬mor cells. This resulted in the inhibition of cell cycle progression at the G0/G1 transmission point with a consecutive increase in apoptosis [9, 11, 20]. In a model of xenografted human pancreatic cancer cells, inhibition of orthotopic tumor growth was achieved most likely by a combination of inhibited mitogenesis, in¬creased apoptosis, and reduced angiogenesis in PD173074-treated animals. This study showed that a high-dose single drug treatment may have clinically relevant therapeutic activity, but data in patients are not yet available.
The concept of creating synergistic antitumor effects by the combined inhibition of several pathways through the use of combination thera-pies [such as by inhibiting the epidermal growth factor receptor (EGFR) and the VEGF-receptor pathways using different drugs] represents an attractive approach. Indeed, clinical studies in patients with advanced colorectal cancer have shown an additive affect of such a combination with regard to response, progression-free sur¬vival, and overall survival using the anti-VEGF bevacizumab and the anti-EGFR cetuximab. This approach has been clinically tested in patients with pancreatic cancer too, but results are not yet available [33].
Using multikinase inhibitors such as sorafenib—thus targeting several pathways, in-cluding VEGF signaling, via a single molecule, and thus showing antiangiogenetic properties in preclinical models—is an alternative treat¬ment strategy to the combination of drugs. The treatment option with sorafenib has been dem-onstrated in preclinical models and in early clinical studies to have activity against several tumor types including adenocarcinoma of the pancreas.
Matrix metalloproteinases (MMP) are dif¬ferent proteolytic enzymes responsible for the breakdown of connective tissue proteins [12]. They play an important role in growth regula¬tion, differentiation, tumor cell spread, and tis¬sue repair. The activity of MMP is highly regu¬lated at different levels. Preclinical and clinical data show that MMP overexpression correlates with increased tumor cell growth and spread in several malignancies [26, 38, 41]. Inhibition of MMP and restoring the normal balance of pro-teolytic activity may prevent tumor growth and metastasis [13, 18, 49]. A low molecular weight MMPinhibitor, marimastat, has been demon-strated to inhibit tumor growth and spread in preclinical cancer models [48, 49] including hu-man pancreatic cancer.
The mammalian target of rapamycin (mTOR) is a serine-threonine kinase with important effects on the regulation of cell growth and proliferation [17]. In a preclinical model of human pancreatic cancer rapamycin alone and, more actively, its combination with an anti-VEGF antibody strongly inhibited primary and metastatic tumor growth [42]. In this in¬vestigation combination therapy improved the effect of single agent treatments. Rapamycin in combination with anti-VEGF antibody inhibited pancreatic tumor cell proliferation, induced apoptosis, and decreased tumor angiogenesis. A clinical trial with rapamycin in pancreatic cancer is ongoing.

14.3    Clinical Studies in Patients
with Pancreatic Adenocarcinoma
There are several different antiangiogenic treat-ment strategies that have been investigated in pa-tients with pancreatic cancer. These include the matrix metalloproteinase inhibitors, agents in-terfering with the VEGF signaling pathway, and several others such as thalidomide, cyclooxygen-ase II inhibitors, and EGFR inhibitors.

14.3.1  Matrix Metalloproteinase Inhibitors
A randomized dose-finding study [5] compar¬ing single agent marimastat, a low molecular weight, broad-spectrum MMP inhibitor, with gemcitabine demonstrated a clear dose-response effect for marimastat, with 1-year survival rates of marimastat at a dose of 25 mg b.i.d. similar to that of gemcitabine, whereas the lower doses (5 and 10 mg b.i.d.) were clearly less effective than gemcitabine.
Another prospective randomized study [6] comparing marimastat (10 mg b.i.d.) in com-bination with gemcitabine to gemcitabine plus placebo in 269 patients with advanced pancreatic cancer failed to demonstrate a significant survival benefit by the addition of marimastat (median survival 165.5 vs 164 days; 1-year survival 18% vs 17%). Grade 3 or 4 musculoskeletal toxicity, a well-known adverse event of marimastat therapy, was reported in less than 5% of the patients.
Due to this study, the further development of marimastat in this indication was stopped.
BAY 12-9566, another MMP inhibitor with
antiangiogenetic properties, is well tolerated and showed antitumor activity in a phase I study in pancreatic adenocarcinoma [19, 22, 24, 25]. A phase III study [36] comparing head-to-head BAY 12-9566 (800 mg b.i.d.) with gemcitabine enrolled 277 patients. In this study the median survival of the MMP inhibitor was demonstrated to be significantly inferior to gemcitabine (3.74 vs 6.59 months).
14.3.2 VEGF Pathway Inhibition
Bevacizumab, a recombinant anti-VEGF mono-clonal antibody, first demonstrated significant improvement in response, progression-free sur-vival, and overall-survival in a phase III random-ized trial in patients with advanced colorectal cancer, when combined with cytotoxic drugs (be-vacizumab 5 mg/kg every second week, together with folinic acid, 5-fluorouracil and irinotecan) [27]. Based on these findings and some evidence of clinical efficacy in pancreatic cancer but with¬out a cancer-specific dose-finding investigation, a phase II study [32] in 52 patients with advanced pancreatic adenocarcinoma combining bevaci-zumab (10 mg/kg every second week) with gem¬citabine as the first-line treatment was initiated. Of the patients, 21% achieved a partial response and another 46% of patients had stable disease resulting in a median progression-free survival of 5.4 months and a median overall survival of 8.8 months—results clearly better than those usually reported for single-agent gemcitabine. Grade 3 and 4 toxicities included hypertension and thrombosis in 19% and 13% of the patients, respectively. Intestinal perforations occured in 8% and bleeding in 2%. There was no correlation observed between pretreatment VEGF plamsa levels and response to anti-VEGF therapy.
Due to these results, a phase III trial [34] of gemcitabine plus bevacizumab as compared to gemcitabine plus placebo was initiated by the cancer and leukemia group B (CALGB) but stopped recruitment after an interim analysis showing that there was no chance of reaching the primary study endpoint of a significant pro-longation of overall survival due to the addition of bevacizumab. Accordingly, the first study re-sults presented at the American Society of Clini-cal Oncology (ASCO)-GI 2007 symposium do not support the superiority of bevacizumab plus gemcitabine: median survival of the 302 patients on bevacizumab and gemcitabine was 5.7 months as compared to 6.0 months for the 300 patients on placebo and gemcitabine.
There is some discussion with regard to the 10 mg/kg of bevacizumab given every 2 weeks used in this trial, as studies in colorectal cancer had demonstrated clinical activity with lower doses (5 mg/kg every 2 weeks) of bevacizumab. On the other hand (interestingly too) there was no increase in gastrointestinal perforations (0% vs 0%) or grade 3/4 bleedings (3% vs 2%) re¬ported for the experimental arm.
Despite the fact that single agent gemcitabine is standard therapy for patients with advanced pancreatic cancer [10], there are several hints in favor of combination chemotherapy in this in-dication too. This is true especially for the com-bination of gemcitabine with platin or fluoropy-rimidine derivates. Premature results of ongoing multicenter phase II studies reported response rates of 11%-22% and median overall survival times of 7.5 to 8.9 months when bevacizumab was added to gemcitabine/oxaliplatin, gem-citabine/cisplatin, or gemcitabine/capecitabine chemotherapy [30, 35, 44]. In these studies se¬vere toxicities with regard to perforations, bleed¬ing, and thromboembolism have been reported. Final study results will be reported soon and may warrant new phase III studies.
Due to the risk of visceral perforation, bleed-ing, and thrombosis, most likely due to the co-medication with bevacizumab, patients with lo¬cally advanced pancreatic cancer with duodenal involvement are excluded in most phase II study of bevacizumab plus radiation therapy in locally advanced nonmetastasized pancreatic cancer. A dose-finding study [14] for bevacizumab to¬gether with capecitabine (825 mg/m2 b.i.d. 5 days a week) and concomitant to radiation therapy with 50 Gy was initiated in patients with locally advanced pancreatic adenocarcinoma. The addi-tion of bevacizumab did not increase the acute toxicity of this radiochemotherapy as compared to historical controls. An overall response rate of 19% was obtained with 6 out of 12 patients, dem-onstrating a partial response at the 5 mg/kg level of bevacizumab.


14.3.3 Other Antiangiogenic Strategies

14.3.3.1 Thalidomide
Thalidomide has shown antitumor activities in several malignancies, especially hematological neoplasms such as multiple myeloma [45]. In ad-dition, in renal cell carcinoma, prostate cancer, and hepatocellular cancer, antitumor effects have been reported [43]. The antitumor effect of tha-lidomide is not clearly understood [16]. In addi-tion to inhibition of tumor necrosis factor alpha, clinical efficacy may be mediated, at least in part, by anti-VEGF effects. A phase I/II clinical study [15] of combination therapy with gemcitabine, celecoxib (400 mg b.i.d.), and thalidomide (200-300 mg o.d.) resulted in no tumor regression, but a mean overall survival of 10 months was reported. Treatment was clinically tolerable with skin rash in 25% of the patients.
Another 50 patients with advanced pancreatic adenocarcinoma who had a weight loss of at least 10% were randomized to receive thalidomide (200 mg daily) or placebo in a single-center double-blind randomized controlled trial [23]. At 4 weeks, 17 patients on thalidomide gained an average of 0.37 kg compared to a loss of 2.21 kg in 16 controls. At 8 week, 12 patients on thalidomide lost 0.06 kg vs 3.62 kg in 8 controls. In this study, data on tumor response are not available.

14.3.3.2    Inhibition of Epidermal
Growth Factor Receptor
EGFR is a well-recognized target of anticancer therapy [2]. Monoclonal antibodies and small molecule tyrosine kinase inhibitors have been shown to be active in different cancers and have obtained approval. Results obtained with this treatment strategy are reported elsewhere in this issue of Recent Results in Cancer Research. Furthermore, the first results of a randomized phase II study [33] comparing gemcitabine/be-vacizumab together with erlotinib or cetuximab suggest an increase in response rate (ca. 20%) by the use of triple drug therapies.
If this holds true will soon become evident, as the results of a major phase III study, the com-bination of gemcitabine/erlotinib together with bevacizumab or placebo, will be reported.


14.3.3.3    Cyclooxygenase 2 Inhibition
Cyclooxygenase (Cox 2) stimulates tumor growth by its proangiogenic and apoptosis-inhibiting ef-fects. Celecoxib, a specific Cox 2 inhibitor, has antitumor activity against several human cancers in preclinical models, including pancreatic can¬cer xenograft [3, 39, 47, 53].
There were 28 patients included in a phase II study [52] evaluating the role of celecoxib in addition to gemcitabine in advanced pancre¬atic cancer. The results of 20 patients were pre¬sented on the annual meeting of the ASCO in 2006 and showed a median overall survival of 6.2 months—not dramatically different from re¬sults expected for gemcitabine alone. Grade 3 or 4 nonhematological toxicities included nausea, vomiting, supraventricular arrhythmias, dys¬pnea, pleural effusions, and hyponatremia, as well as gastrointestinal bleeding in one patient.
Further evaluation of this agent has been stopped due to cardiac toxicity possibly caused by celecoxib [7].

14.4    Conclusions
Adenocarcinoma of the pancreas remains a major cause of cancer morbidity and mortal¬ity worldwide. Recent results confirm that the dismal prognosis of patients can be improved by small steps. Still, there is an urgent need for novel treatment strategies. In contrast to anti-EGFR targeted, small-molecule therapy, the first large clinical trials failed to prove the effectivity of other antiangiogenic strategies in combina¬tion with gemcitabine in this indication. Never¬theless, stimulated by the results with erlotinib, antiangiogenic treatment options continue to be among those most likely to further improve the prognosis in this tumor entity.
 

Surgical Palliation of Advanced Pancreatic Cancer


In about 80% of patients with pancreatic cancer surgical resection is not feasible at the time of diagnosis. Therefore, palliative treatment plays a key role in the treatment of pancreatic cancer. The defined goals of palliative treatment are: reduc-tion of symptoms, reduction of in-hospital stays, and an adequate control of pain. In patients with nonresectable pancreatic carcinoma the leading goal of palliative strategies should be the con¬trol of biliary and duodenal obstructions such as jaundice-associated pruritus or sustained nausea and vomiting due to gastric outlet obstruction. Although the role of endoscopy for palliation has been increasing, operative palliation is still indicated in selected cases. Obstructive jaundice is found in approximately 70% of patients suf-fering from carcinoma of the pancreatic head at diagnosis and has to be eliminated to avoid progressive liver dysfunction and liver failure. In up to 50% of patients with pancreatic cancer, clinical symptoms such as nausea and vomiting occur. For the treatment of malignant biliary ob-structions in patients with pancreatic carcinoma, endoscopic biliary drainage is the option of first choice. In case of persistent stent-problems such as occlusion or recurrent cholangitis, a hepatico-jejunostomy should be considered. The role of a prophylactic gastroenterostomy is still under dis¬cussion. In patients with combined biliary and gastric obstruction a combined bypass should be performed to avoid a second operation. The sig¬nificance of laparoscopic biliary bypass is not yet clear. A surgical, minimally invasive approach for treating bile duct obstruction is not the stan¬dard nowadays. The role of surgical pain relief is mostly negligible today. Computed tomography (CT)- or EUS-guided celiac plexus neurolysis has replaced surgical intervention today. The significance of palliative resections is currently a controversial topic. However, beyond controlled randomized studies, a palliative pancreaticoduo-denectomy in patients with advanced pancreatic carcinoma cannot be recommended at this time.

13.1    Introduction
Considerable advances in the treatment of pa-tients with pancreatic cancer have been reached during recent decades and surgical results after pancreatic head resection have clearly improved in the majority of patients, yet the disease is di-agnosed too late for a curative surgical approach [1]. This fact and the aggressiveness of the pan-creatic adenocarcinoma are the reasons for the poor overall 5-year survival rate, which has only moderately increased from less than 5% to ap-prox. 7% nowadays. In about 80% of the patients coming to diagnosis with pancreatic carcinoma, palliative therapy is the only treatment option.
Palliative strategies in patients with pancre¬atic carcinoma focus on three symptoms: pain, duodenal obstruction, and obstructive jaundice, whereby the palliative treatment of these symp-toms is primarily directed at reducing the clini¬cal symptoms, reducing the hospital stay, and last but not least ensuring as much overall quality of life as possible. Currently both nonoperative endoscopic procedures and surgical techniques are available to provide palliation of the leading symptoms, and the principal goal of a palliative treatment plan should be tailored to most effec¬

tively suit the patients' clinical presentation and their overall physical and mental condition; it should also incorporate the estimated prognosis.
In the past, surgery has been the only treat-ment option for effective palliation in patients with pancreatic carcinoma. But with the rapid advances in the development of endoscopic tech-niques, the significance of surgical palliation has declined. The main consideration for avoiding surgical palliation is the morbidity and mortal¬ity associated with surgical procedures such as gastroenterostomy, hepaticojejunostomy, and even laparotomy, although the latter occurs only in advanced cancer patients. It remains, however, unclear as to how the value of surgical palliation vs endoscopic palliation may be appropriately judged, and neither life quality investigations nor purely outcome-centered evaluations have so far succeeded in establishing a useful therapeutic al-gorithm for this severely ill patient population. Hence, decision-making on a palliative strategy for an individual patient remains difficult and may have to be based on an interdisciplinary dis-cussion between the patient, oncologist, thera-peutic radiologist, and surgeon.

13.2    Palliative Therapy of Biliary
Obstruction: Stent or Surgery?
The surgical options for palliative therapy in biliary obstruction include operative placement of biliary drains such as a T-drainage, choledo-choduodenostomy, hepaticojejunostomy, or, in rare cases, a peripheral or distal hepaticojejunos-tomy (Fig. 13.1). It is important to note that the often-discussed argument that the placement of a T-drainage is a very small surgical procedure with a low complication rate is, on one hand, correct; however, it has to be considered that this procedure creates an external biliary fistula with all its possible complications and implications for life quality. It should therefore be considered only when other measures of palliative treatment of obstructive jaundice have failed or cannot be undertaken for technical reasons. In addition, the amount of bile loss through such an external biliary fistula may lead to a profound electrolyte and fluid imbalance; hence, in such cases we ad-vocate a simple anastomosis between jejunum and the gallbladder to provide relief of biliary obstruction. The cholecystogastrostomy de¬scribed in earlier reports may lead to bile gastri¬tis, increased gastrin release, and secondary acid hypersecretion, as well as food entry into the bili-ary system and subsequent recurrent obstruction or cholangitis (or both) [2]. We therefore have abandoned this procedure along with most other centers experienced in pancreatic surgery.
Hence, the cholecystojejunostomy remains the standard surgical procedure for palliation when the surgical dissection of the hepatoduo-denal ligament has to be avoided. If cholecys-tojejunostomy is chosen, the cystic duct ought to attach a common bile duct and the distance to the tumor mass needs to be at least 2-3 cm to prevent early reobstruction by continuing tumor growth. Several trials have compared cholecystojejunostomy and hepatic enterostomy to evaluate whether the risk of bile duct injuries due to resection of the hepatoduodenal ligament may be avoided. Watanapa et al. [36] found that cholecystoenterostomy yielded a success rate of 89%, which was not significantly different from a success rate of 97% in patients receiving a hepaticoenterostomy. In addition, the authors found that cholangitis and recurrent jaundice were observed in 20% of the cholecystojejunos-tomy cases, whereas the complication rate in the group with hepaticoenterostomy was higher. The authors concluded that there may be a slightly increased risk of surgical complications when dissecting the hepatoduodenal ligament for he-paticojejunostomy. Furthermore, other authors have indicated that the possible troublesome dissection of the hepatoduodenal ligament may often be avoided when the common bile duct is transected in the middle or lower section and a side-to-side choledochoenterostomy is performed rather than the standard end-to-side hepaticojejunostomy.
Some authors have evaluated the choledocho-duodenostomy, which has been proved to be an effective surgical method for treating obstructive jaundice in benign conditions and also has been used in selected cases for biliary reconstruc¬tion after orthotopic liver transplantation [3]. However, in the case of pancreatic carcinoma, which has led to jaundice, many surgeons today feel that, in the advanced stages of pancreatic carcinoma (when patients are receiving pallia¬tive surgical treatment), an anastomosis in close proximity to the tumor may lead to early reste-nosis and occurrence of jaundice. Additionally, the peritumoral inflammation usually leads to stiff duodenum, which will not allow attention-free anastomosis, thereby increasing the risk for anastomotic leakage. However, other authors have utilized the choledochoduodenostomy rou¬tinely and have shown that the procedure is as¬sociated with a lower complication rate, a short length of postoperative hospital stay, and a very low recurrence rate of obstructive jaundice (be¬low 2%). Therefore, since the overall complica¬tions rate for the other methods of biliary bypass were higher, they advocate the choledochojeju-nostomy as the standard method for surgical palliation in obstructive jaundice caused by ad¬vanced pancreatic carcinoma [4].
The introduction and development of endo-scopic methods of biliary reconstruction reach¬ing from papillotomy to placements of intra-ductal stents in patients has revolutionized the palliative treatment of patients with obstructive jaundice due to pancreatic cancer. Today endo¬scopic placement of biliary stents is accepted as a standard treatment in patients with unresectable pancreatic carcinoma. However, the controversy regarding the abdominal palliative treatment— stent or surgery—is still ongoing and undecided. Several prospective randomized trials have com¬pared nonoperative biliary stenting with opera¬tive procedures such as hepaticojejunostomy or others. The study by Shepherd et al. did not show a significant difference in complication rate, 30-day mortality rate, incidence of postoperative gastric outlet obstruction, or median survival [5]. However, the rate of recurrent jaundice was significantly higher after biliary stenting com-pared to the surgical bypass procedure (43% vs. 0%). Furthermore, in a randomized trial Smith et al. demonstrated that recurrent jaundice oc¬curred more often in patients after stent place¬ment than in patients after surgical biliary bypass [6] (Table 13.1).
The main argument for surgical bypass is that the surgical procedure is thought to be a defini-tive treatment avoiding the regular endoscopic procedures for changing of stents or treating stent complications, which are frequent in this patient population. In addition, many surgeons feel that the definitive palliative surgical procedure is more cost-effective for the same reason. However, a re¬cent study by Artifon and coworkers shows that endoscopic biliary drainage carries lower costs and provides better quality of life when compared to palliative surgical procedures [7]. Again, as in most other studies comparing surgical endo-scopic bypass, no difference in the median sur¬vival of the investigated groups was found.
Taken together it is still unclear whether there is a standard treatment algorithm to be advo-cated, since all studies carry the problem of bias in patient selection and the lack of acceptable and validated quality-of-life data. Nevertheless, endoscopic biliary drainage has become the gold standard for palliation of malignant bile duct ob-structions in patients with pancreatic carcinoma, and the numbers of surgical palliative procedures have clearly declined. However, surgical options still carry significance. For example, in the case of refractory stent problems such as stent occlusion or recurrent cholangitis, operative stent with¬draw and hepaticojejunostomy may be indicated. Furthermore, primary hepaticojejunostomy should be performed in cases of endoscopically impassable tumor masses; finally, if an advanced pancreatic tumor is judged to be nonresectable at laparotomy, a prophylactic hepaticojejunos-tomy should be considered in patients with ob¬structive jaundice or in the case of threatening obstructive jaundice. To decide on the optimal treatment strategy for a patient with biliary ob¬struction due to pancreatic carcinoma, a close collaboration between the surgeon, the endos-copist, and the oncologic specialist is necessary; complicated cases should be managed based on interdisciplinary approaches.

13.3    Palliative Surgery for Gastric Outlet Obstruction Alone or in Combination with Biliary Bypass?
The standard palliative surgical procedure for gastric outlet obstruction due to upper abdomi-nal malignancies is a retro- or antecolic end-to-side or side-to-side gastrojejunostomy. While  this is normally a simple surgical procedure due to the often marginal clinical condition of the patients in advanced tumor stages, the gastroje-junostomy shows high morbidity and mortality rates [8]. The question of whether a prophylac¬tic gastroenterostomy is rational and should be performed when a normal resectable situation in patients with pancreatic head carcinoma is found at laparotomy is undecided as of yet and under discussion. A recent study by Egrari et al. shows that the mean time to obstruction was 15.7 months compared to a mean overall survival of approx. 13 months in patients with advanced pancreatic carcinoma. The authors demonstrated that due to the rapid natural progression of pan¬creatic adenocarcinoma, most patients do not survive long enough to obstruct and therefore do not need a prophylactic gastroenterostomy
[9].
Today many investigators feel that, due to possible morbidity and mortality, prophylac¬tic gastroenterostomy is unnecessary, and only a selective use of gastroenterostomy should be exercised in the case of present or impending duodenal obstruction that has already led to clinical symptoms. A second area of discussion is the question whether a combination of bili¬ary and gastric bypass is reasonable and profit¬able for the patient with pancreatic carcinoma in the palliative situation. A French study analyzing 2,493 patients with unresected cancer of the pan¬creas demonstrated that the mortality in patients with a combination of biliary and gastric bypass was similar [10]. However, they also observed that 16% of the patients undergoing biliary by¬pass alone developed a gastric obstruction. This finding was confirmed by other groups [11, 12]. Therefore, a number of authors concluded that a combination of biliary and gastric bypass as the initial procedure should be performed, since it minimizes the risk of reoperation and provides definitive palliation [10, 13]. To create a gastro-jejunostomy in addition to a surgical bypass of biliary obstruction is not a technical challenge for experienced general surgeons and today is as¬sociated with low morbidity and mortality rates. However, the decision for an initial combination of biliary and gastric bypass depends on several factors such as preexisting gastric outlet obstruc¬tion at the time of operation, imminent gastric obstruction, the overall condition of the patient, tumor stage, and tumor biology (Fig. 13.2).
As stated above, the best therapeutic strategy and the surgical method chosen for an individual patient should be discussed with consideration for all clinical factors defining the individual patient; when surgical options are considered, it seems important to underline that for patients with unresectable pancreatic cancer who present clinically manifest gastric obstruction at admis-sion, the median survival often may be as little as 4 weeks, even when newer oncologic treatment concepts are initiated [14]. This may be an im-portant argument for an initial combined biliary and gastric bypass to ensure that such patients have the chance to leave the hospital with imme-diately effected palliation.

13.4   Minimally Invasive Procedures for Surgical Palliation
Throughout the last few decades, minimally in-vasive procedures for palliative surgery have been reported in increasing numbers. This holds true also for palliative biliary and gastric bypass procedures, and today a considerable number laparoscopic gastric and biliary bypasses for periampullary carcinomas have been reported in the literature [15, 16]. In these studies, lapa-roscopic techniques are either performed as cho-lecystojejunostomies or hepaticojejunostomies to create a biliary bypass. The possible advan¬tages of minimally invasive surgical approaches seem obvious since especially in those severely ill patients the trauma of the surgical procedure and the time of hospital stay are very important factors. However, some possible disadvantages of the minimally invasive procedures have to be considered. The mean operating time seems to be significantly longer compared to standard open surgical procedures, thereby increasing the surgical trauma. Furthermore, special laparo-scopic expertise is required to ensure a low com¬plication rate, since both a hepaticojejunostomy and gastroenterostomy are considered advanced laparoscopic procedures. Today, endoscopic stenting via endoscopic retrograde cholangiogra-phy is the gold standard for palliation in patients with malignant bile duct obstruction due to car¬cinoma of the pancreatic head, and although several authors reported results for laparoscopic biliary bypass in single patients, no prospective randomized study comparing laparoscopic sur¬gery vs stenting has yet been reported. The first choice of treatment in patients with bile duct obstruction due to pancreatic cancer should therefore be the endoscopic stenting. If indicated (based on repeated stent occlusions or recurrent cholangitis), surgical intervention regardless of the surgical technique should be discussed. Dur¬ing this discussion it has to be considered that the laparoscopic biliary bypass is not a standard minimally invasive procedure and only experts in the field of laparoscopy should perform such operations in this very ill patient population.
In a small group of patients, Kazanjian et al. demonstrated that laparoscopic gastrojejunos-tomy is a safe and effective palliation for patients with gastric outlet obstruction due to pancreatic carcinoma. In their analysis it was especially sig¬nificant in a group of patients with a very limited survival [17]. In addition, a group from Norway compared open vs laparoscopic gastrojejunos-tomy for palliation in advanced pancreatic can¬cer retrospectively and found that laparoscopic gastrojejunostomy in advanced cases offered a reduced estimated blood loss and a shortened hospital stay when compared to open gastrojeju-nostomy [18]. Hence, at this time minimally in¬vasive procedures using standardized techniques should be considered for relief of gastric outlet obstruction due to pancreatic carcinoma when the laparoscopic expertise is present. In this situ¬ation a low complication rate can be ensured and the minimally invasive techniques might be a vi¬able alternative for open surgical procedures, es-pecially in patients with a very limited prognosis
[19].

13.5    Role of Surgical Pain Relief
The surgeons treating patients with advanced pancreatic head carcinoma have to keep in mind that quality of life is the most important factor for these patients who have such a dire prognosis. In this context, pain is the most feared symptom for a majority of the patients and for many of them pain constitutes a clinically significant problem until death; pain management is troublesome.
Pain fibers from the pancreatic gland (the ce-liac ganglion) run within the major and minor splanchnic nerves to the spinal column. An in-terruption of this pathway can provide pain re¬lief, and such a disruption can be accomplished by either targeting the abdominal or the thoracic cavity. The first intraoperative chemical splanch-nicectomy was introduced by Copping and col¬leagues in 1969 [20]. In their clinical experience reported almost 10 year later, approximately 90% of patients with pain at diagnosis experienced significant relief after intraoperative chemical splanchnicectomy [21]. Since then many in¬vestigators have utilized this method, and Lil-lemoe and coworkers reported in a randomized controlled trial that intraoperative chemical splanchnicectomy with 50% alcohol significantly reduced or prevented pain in patients with unre-sectable pancreatic cancer [22]. In contrast, van Geenen and coworkers from Amsterdam could not confirm these findings. In their randomized study, patients were divided into three groups: (1) palliative bypass surgery receiving intraop-erative celiac plexus blockade, (2) palliative by¬pass surgery without celiac plexus blockade but followed by high-dose conformal radiotherapy, and (3) palliative bypass surgery with both (ce-liac plexus blockade, followed by high-dose con-formal radiotherapy). They concluded that celiac plexus blockade for pain management did not result in an increase to pain medication-free sur¬vival and therefore presumed that celiac plexus blockade could not demonstrate a positive effect on pain management for the patients with ad¬vanced pancreatic carcinoma [23]. To disrupt the pain neuropathway, splanchnic nerves within the thorax could also be interrupted. This can be ac-complished either via thoracotomy or via video-assisted thoracoscopy (VATS) [24, 25]. In recent times, the plexus blockade has been reached via nonsurgical interventions, namely using endo-scopic ultrasound (EUS) or wild-guided tech¬niques. A prospective study of the EUS-guided celiac plexus neurolysis for pain treatment in patients with advanced pancreatic head can¬cer showed that the technique is safe and yields pain control [26]. In light of such nonsurgical alternatives for celiac plexus blockage, the role of surgical pain relief seems to be marginal nowa¬days. However, in selected patients, namely who are not responding to noninvasive methods of plexus blockade, a surgical intervention may still be indicated.

13.6    Palliative Resection: Does It Play a Role?
Adenocarcinoma of the pancreatic head is con-sidered one of the gastrointestinal malignancies with the worst prognosis. If no standardized op-erative procedures have been established (classi-cal Kausch-Whipple resection, pylorus preserv¬ing pancreatic head resection) the overall 5-year survival rate of patients with pancreatic head carcinoma today is estimated to be approx. 5%, and the 5-year survival rate after curative resec-tion reaches approx. 20% in specialized centers around the world [27, 28]. However, differing from earlier reports, the perioperative mortal¬ity has decreased significantly during the last few decades, and today morbidity rates around 15% and mortality rates below 3% for standard¬ized pancreatic head resection have been reached in high-volume centers. This fact and the con-sideration that the patient with pancreatic head carcinoma in the majority of cases presents in stage 3, in which advanced disease is present and undetected further tumor spread has to be ex-pected, many investigators today believe that real curative resections are rare events. Further argu-ments on this line are supported by the results of recent multicenter trials in adjuvant chemother-apy after R0 or R1 resections for pancreatic head carcinoma showing that even the patients with R1 resection profit considerably from postopera¬tive adjuvant therapy [29]. Therefore, oncologists have long proposed that the pancreatic head re-section for a defined tumor no longer be termed a curative resection, since in most of the cases advanced stages of the disease are present. Many investigators today believe that all resections for pancreatic head carcinoma are, in principle, pal-liative. Following this argument, the goal of the resection may change much more toward reach¬ing quality of life for the patients, as with other palliative procedures.
At present no prospective data are available in which a palliative resection was investigated in a randomized fashion. Several authors have re-ported about retrospective data comparing pal-liative procedures (biliary and gastric bypasses combined or alone) with pancreatic resections [30]. Lillemoe et al. investigated the role of pallia-tive resection compared to combined biliary and gastric bypass, showing a significant improved overall survival for patients undergoing pallia¬tive pancreaticoduodenectomy. All patients were patients in which, after transecting the pancreas (passing the point of no return during pancreatic head surgery), nonresectability was found in the retro-pancreatico-duodenal plane [31]. However, in this study no further subdivisions regard¬ing the R-status was accomplished. In another study, Reinders et al. compared patients after a microscopically nonradical pancreaticoduode-nectomy with patients after surgical bypass [32]. Both studies neither evaluated the quality of life nor the long-term follow-up criteria and only showed that the so-called palliative pancreati-coduodenectomy procedures yield significantly better results and longer survival than ones in which patients received surgical bypass, leaving their primary tumor mass in place. In one recent study the investigators compared patients in a palliative situation undergoing double loop by¬pass surgery with patients undergoing palliative pancreaticoduodenectomy. Special emphasis was laid on the investigation of quality of life in this study. All patients undergoing bypass were sub-grouped into those with locally advanced disease and those with metastasized diseased. The 1-year survival was 25% in the palliative resected group vs 20% in the locally advanced and 15% in the metastasized disease group. The quality-of-life data were favorable for the patients after bypass surgery; however, the morbidity and mortality rates in patients after palliative resection were elevated [33].
These results prompted us to propose a study in which the role of the palliative resection it¬self should be evaluated. Following extensive interdisciplinary discussions with gastroenter-ologists and oncologists, we derived a protocol in which patients with carcinomas of the pan¬creatic head that had already metastasized into the liver at diagnosis and revealed a resectable situation were randomized into two groups. One group would receive standard gemcitabine che¬motherapy until tumor progression, whereas the other group would receive a pancreatic head re¬section with or without liver resection and sub¬sequently standard gemcitabine treatment until tumor progression. The liver resection was only to be performed when resections could provide a significant tumor mass reduction because the additional surgical risk to the pancreatic head procedure was to be avoided This study is now underway (and hopefully open for recruitment) and we will be able to analyze the results in the near future.
Palliative pancreatic head resections outside of accepted study protocols should not be per-formed since the significant additional clinical risk of complications for morbidity and mortality is not acceptable; they must only be performed in the framework of randomized prospective trails.

13.7    Summary
For the treatment of malignant biliary obstruc-tions in patients with pancreatic carcinoma, en-doscopic biliary drainage is the option of first choice. In case of persistent stent-problems such as occlusion or recurrent cholangitis, a hepatico-jejunostomy should be considered. The role of a prophylactic gastroenterostomy is still under dis¬cussion. In selected patients with duodenal ste¬nosis present at the time of operation, or patients with impending duodenal obstruction, a prophy¬lactic gastroenteric bypass may be indicated. The same should be considered for patients showing a duodenal stenosis during an operation for biliary obstruction. In such patients an initial combined biliary and gastric bypass should be performed to avoid a second operation for gastric outlet ob¬struction. The significance of laparoscopic bili¬ary bypass is not yet clear. A surgical, minimally invasive approach for treating bile duct obstruc¬tion is not the standard nowadays, and it should be reserved for experts in the field of laparos-copy. Otherwise, laparoscopic gastrojejunostomy is a standardized surgical procedure that offers significant advantages in regards to morbidity and mortality compared to open surgical tech¬niques. The role of surgical pain relief is mostly negligible today. Computed tomography (CT)-or EUS-guided celiac plexus neurolysis have re¬placed surgical interventions. The significance of palliative resections is a controversial topic now¬adays. However, beyond controlled randomized studies, a palliative pancreaticoduodenectomy in patients with advanced pancreatic carcinoma cannot be recommended at this time.