All.dvi

Chapter 2
Metastasis and Drug Resistance

Dominic Fan, Sun-Jin Kim, Robert L. Langley, and Isaiah J. Fidler
Multidrug resistance (MDR) phenotype emerging from chemotherapy is a major problem in managing patients with metastatic cancers. The discovery thata cardiovascular drug, verapamil, can bind to P-glycoprotein and reverse MDR ini-tiated serious research efforts in MDR-reversal by various compounds and modesof pharmacological modifiers. Those include major calcium channel blockers suchas bepridil, diltiazem, felodipine, isradipine, nicardipine, nifedipine and nimodip-ine, verapamil and analogs; calmodulin antagonists; antibiotics and analogs; indolealkaloids; cyclosporins and analogs; hormones and antihormones; pharmaceuticalemulsifying surfactants; liposomal encapsulation; etc. The majority of the studiestargeted one of the MDR mechanisms, P-glycoprotein. These studies have beensuccessful under in vitro and limited in vivo animal conditions; the correlations forclinical trails are still lacking. Therefore, an effective MDR-reversing chemother-apy is not available. It is the purpose of this chapter to review the past and currentexperimental reversal of MDR and, in particular, the importance in targeting drugresistance in relevant cancer metastasis models.
Keywords Metastasis · MDR · Apoptosis · Animal models
Despite improvements in diagnosis, surgical techniques, patient care, and adjuvanttherapies, most deaths from cancer are due to metastases that are resistant to conven-tional therapies (Fidler 1990). The major obstacle to effective treatment is tumor cellbiologic heterogeneity. Moreover, the metastases can be located in different organs,and the specific organ environment can influence the biologic behavior of metastatic D. Fan (B)Department of Cancer Biology, Cancer Metastasis Research Center, Unit 854, The University ofTexas M. D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USAe-mail: [email protected] K. Mehta, Z.H. Siddik (eds.), Drug Resistance in Cancer Cells, DOI 10.1007/978-0-387-89445-4 2, C  Springer Science+Business Media, LLC 2009 D. Fan et al.
cells, including their response to systemic therapy. Only a better understanding ofthe molecular mechanisms that regulate the process of metastasis and the interac-tions between the metastatic cells with the organ microenvironment can provide afoundation for the design of more effective therapy.
The Pathogenesis of Metastasis
The process of metastasis is highly selective and consists of a series of sequential,interrelated steps. To produce clinically relevant lesions, metastatic cells must com-plete all steps of this process. After the initial transformation and growth of cells,vascularization must occur if a tumor mass is to exceed 1 mm in diameter. The syn-thesis and secretion of several proangiogenic factors by tumor and host cells and theabsence of antiangiogenic factors play a key role in establishing a capillary networkfrom the surrounding host tissues. Next, local invasion of the host stroma occursas a consequence of the enhanced expression of a series of enzymes (e.g., collage-nase). Once tumor cells penetrate lymphatic or vascular channels, they may growat the invasion site or detach and be transported within the circulatory system. Thetumor emboli must survive immune and nonimmune defenses and the turbulenceof the circulation, then arrest in the capillary bed of receptive organs, extravasateinto the organ parenchyma, proliferate, and establish a micrometastasis. Growth ofthese microscopic lesions requires development of a vascular supply and evasion ofhost defense cells. When the metastases grow, they can shed tumor cells into thecirculation to produce metastasis of metastases (Fidler 1990).
The outcome of the metastatic process depends on multiple and complex interac- tions of metastatic cells with host homeostatic mechanisms (Fidler 1997). More thana century ago, Stephen Paget researched the mechanisms that regulate organ-specificmetastasis, i.e., pattern of metastasis by different cancers, and questioned whetherthe organ distribution of metastases produced by different human neoplasms wasdue to chance and analyzed more than 700 autopsy records of women with breastcancer. His research documented a nonrandom pattern of visceral (and bone) metas-tasis. This finding suggested to Paget that the process was not due to chance but,rather, that certain tumor cells (the "seed") had a specific affinity for the milieu ofcertain organs (the "soil"). Metastases resulted only when the seed and soil werecompatible (Paget 1889).
A current definition of the "seed and soil" hypothesis consists of three principles.
First, neoplasms are biologically heterogeneous and contain subpopulations of cellswith different angiogenic, invasive, and metastatic properties (Fidler 2003; Lang-ley and Fidler 2007). Second, the process of metastasis is selective for cells thatsucceed in invasion, embolization, survival in the circulation, arrest in a distant cap-illary bed, and extravasation into and multiplication within the organ parenchyma.
Although some of the steps in this process contain stochastic elements, as a whole,metastasis favors the survival and growth of a few subpopulations of cells that pre-exist within the parent neoplasm (Fidler and Kripke 1977; Talmadge et al. 1982).
2 Metastasis and Drug Resistance Thus, metastases can have a clonal origin, and different metastases can originatefrom the proliferation of different single cells (Fidler and Talmadge 1986; Hu et al.
1987; Talmadge et al. 1982).
Third, and perhaps the most important principle for the design of new can- cer therapies, is that the outcome of metastasis depends on multiple interactions("cross-talk") of metastatic cells with homeostatic mechanisms, which the tumorcells can usurp (Fidler 1995). Therapy of metastasis, therefore, can be targeted notonly against tumor cells but also against the homeostatic factors that promote tumorcell growth, survival, angiogenesis, invasion, and metastasis.
One of the most depressing and predictable facts of cancer management is the devel-opment of the multidrug resistance (MDR) phenotype in patients treated chronicallywith certain natural chemotherapeutic drugs. This clinical phenomenon accounts forthe unsatisfactory low incidence of response rate for the majority of solid tumors tochemotherapy – one of the few conventional treatments for metastatic diseases inpast few decades.
Since the heterogeneous nature of tumor and cancer metastasis was conceptu- alized (Paget 1889; Fidler 1973; Fidler and Kripke 1977; Fidler 1978; Poste andFidler 1979; Fidler and Poste 1985; Fidler 2001), it is clear that MDR is a resul-tant clinical outcome manifested by successful cancer cells endowed with multi-ple mechanisms for survival. Like other vital traits of metastatic cancer cells, MDRshould be conceived as a phenotype marked by a collection of independent or collat-eral modifications, overexpressions, and/or amplifications of endogenous moleculesthat interplay with distinct normal cellular pathways (Fig. 2.1) that include ABC Transporters Tubulin Mutation Episomes Amplification Topoisomerase II Mutation Altered Cellular Calcium Levels Enhanced Sodium Pump Activity Altered Reduction-Oxidation Pathways Formation of Double Minute Chromosomes Overexpression of Cytoplasmic 22 kDa Sorcin Fig. 2.1 MDR-associated mechanisms
D. Fan et al.
P-glycoprotein (Juliano and Ling 1976; Kartner et al. 1983), protein kinase-C (PKC)overexpression (Fan et al. 1992a, b; Aftab et al. 1994), ABC transporters (Adachiet al. 2007), tubulin mutation (Inaba et al. 1987), episomes amplification (Ruiz et al.
1989), formation of double minute chromosomes (Von Hoff et al. 1990), alteredcellular calcium levels (Nair et al. 1986), topoisomerase II mutation and alteredreduction–oxidation (Deffie et al. 1988), and the overexpression of cytoplasmic 22-kDa sorcin (Hamada et al. 1988). Those cancer cells employing single and espe-cially unique oncogenic mechanism, presumably exist, would likely be eliminatedby the host defense mechanisms during the progression of cancer or by conventionalcancer therapy and would be without further clinical manifestation. Therefore, ini-tial treatment with chemotherapeutic drugs and subsequent reversal of MDR shouldbe combined as a standard protocol for effective chemotherapy at the onset of cancermanagement, rather than resolving to salvage therapies – when the patients returnwith compromised performance status and growth of refractory tumors.
Unfortunately, an effective MDR-reversing chemotherapy is not available. Since the inception of an organized drug-screening program (DeVita et al. 1979), researchefforts have been largely compound-oriented (sensitive drug-screens) (Frei 1982;Venditti 1981) rather than disease-oriented (tumor panels) (Alley et al. 1988),metastasis-oriented (orthotopic animal models) (Wilmanns et al. 1992; Singh et al.
1994; Killion et al. 1999; Langley and Fidler 2007) or MDR-oriented (relevant resis-tant drug-screens) (Mickisch et al. 1991a, b, Dong et al. 1994). In the followingsections, we review the past and current experimental reversal of MDR and, in par-ticular, the importance in targeting drug resistance in cancer metastasis.
Reversal of Experimental MDR
The majority of the MDR-reversing studies were in vitro assays that cannot addressthe complexity of physiology and pathology in cancer patients, and in particularmetastasis of the cancer. It is physiology and pathology that modulate the progres-sion of cancer and metastasis and the pharmacokinetics (what the host and cells doto the drugs) and pharmacodynamics (what the drugs do to the host and cells) (Fordand Hait 1990). Decisively, the validity of an in vitro assay is governed by its abil-ity to derive an acceptable level of sensitivity (the prediction of true positives) andspecificity (the prediction of true negatives) (Fan et al. 1985).
Calcium Channel Blockers
Verapamil and Other Clinically Approved Agents
As the elements of time and costs stacking up against the development of newanticancer drugs, the initial observation of Tsuruo et al. (1981) of a reversal byverapamil (a coronary vasodilator) on an MDR phenotype in P388 leukemia cells(Tsuruo et al. 1981, 1982) inscribed the beginning of an explosive search for 2 Metastasis and Drug Resistance MDR-reversing anticancer therapeutics. Several major calcium channel blockersapproved for clinical use in the United States were candidates for such a search:bepridil (a pyrrolidylamine), diltiazem (a benzothiazepine), felodipine, isradipine,nicardipine, nifedipine and nimodipine (dihydropyridines), and verapamil (a ben-zeneacetonitrile). Although one of the major biological effects of verapamil is theblockage of the slow-channel-mediated calcium entry into cardiac cells (Kohlhardtet al. 1972) and drug-resistant cancer cells (Bucana et al. 1990), its MDR-reversingmechanism is not clearly understood and may be quite apart from its physiologicaction, in which a trial depolarization plays an important role and is related tothe fast-channel effect for sodium influx (Rougier et al. 1969). Verapamil-mediatedenhancement of intracellular accumulation of MDR-linked anticancer drugs is uni-versally observed and attributed to an effect on P-glycoprotein-mediated efflux ina variety of cancer cell lines (Tsuruo et al. 1982; Inaba et al. 1979; Harker et al.
1986). Extensive efforts were made to identifying and translating the unique MDR-reversing properties of verapamil and other calcium channel blockers into clini-cal terms (Slater et al. 1982; Tsuruo et al. 1983a, b; Fojo et al. 1985; Fine et al.
1987; Ford et al. 1990; Fan et al. 1994a, b). Unfortunately, the adverse hemato-dynamic effects of verapamil have limited its clinical potential for routine use inadjunct chemotherapy (Ozols et al. 1987). In addition to verapamil, many clinicallyapproved calcium channel blocker were shown to affect intracellular accumulationof MDR-linked anticancer drugs and to reverse MDR phenotype in vitro (Tsuruoet al. 1983a, b; Schuurhuis et al. 1987; Hollt et al. 1992; Fan et al. 1994a, b). How-ever, with the exception of the bepridil studies, most preclinical studies employedconcentrations of calcium channel blockers much higher (to achieve experimentalMDR-reversing activity) than the peak plasma levels derived from patients whoseperformance status was less compromised than those of cancer patients enteringclinical trials with advanced disease. Therefore, it was not surprising that difficultieswere encountered in clinical trials using verapamil (Ozols et al. 1987) and with othercalcium channel blockers for reversing drug resistance to standard chemotherapeu-tics in advanced cancer patients. At a micromolar dose range of verapamil, the cyto-toxic effects to normal cells are remarkably severe (Lampidis et al. 1986). Further-more, the high-dose requirement for reversal of MDR in vitro suggested additionaleffects (mechanisms of action) other than a simple physiologic blockage of the cal-cium channels (Huet and Robert 1988). Several groups have shown that verapamilcan bind to P-glycoprotein and compete for binding sites for MDR-related agentsto P-glycoprotein (Cornwell et al. 1987; Safa et al. 1987; Akiyama et al. 1988;Beck et al. 1988). It was shown that in the process of reversing an MDR pheno-type, verapamil also stimulated marked ultrastructural changes (an MDR-associatedtwofold increase in the number of intramembrane particles) of drug-resistant P388cells (Garcia-Segura et al. 1992). Moreover, under certain experimental conditions,treatments of the drug-resistant human colon LS 180 with verapamil, nifedipine,nicardipine, or diltiazem could increase mdr-1 mRNA expression and induce celldifferentiation (Herzog et al. 1993).
If one examines the bulk of in vitro literature and the clinical pharmacoki- netic information, one finds in general a lack of consideration for controlled D. Fan et al.
pharmacokinetic parameters (e.g., plasma elimination half-life of chemosensitizersand of standard anticancer drugs) to simulate relevant pharmacodynamic effects invitro. As nonphysiologic as in vitro assays are, chemical–cell interactions do fol-low the law of concentration and time; this kind of pharmacologic considerationmay help in reducing the frequency and costs in deriving false-positive experimen-tal new drugs and MDR-reversing agents. Therefore, while it may be feasible toseek enhancement for the efficacy of standard anticancer drugs by verapamil andother calcium channel blockers, the selectivity, scheduling, and dose intensity of thechemosensitizers must be taken into consideration inasmuch as these parametersmay influence MDR, tumor spread, and clinical outcome of therapy.
Verapamil Derivatives and Other Experimental Calcium Channel Blockers
The disappointment of the initial clinical trials with verapamil (Ozols et al. 1987)stimulated an intense effort to develop chemosensitizers that were less cytotoxic tonormal cells: one of the critical parameters in systemic cancer therapeutics (Lam-pidis et al. 1986; Fan et al. 1988). A number of structural analogs to verapamil(e.g., devapamil, emopamil, gallopamil, D528, D595, D792) have been implicatedin the reversal of MDR in vitro (Pirker et al. 1990) with marginal toxicity inanimal models (Nawrath and Raschack 1987; Pirker et al. 1989). The less cal-cium antagonistic and less cardiotoxic R-enantiomer of verapamil (versus that ofclinically approved racemic verapamil) could reverse an MDR phenotype in vitro(Mickisch et al. 1990a, b). R-enantiomer of verapamil decreased the expression ofP-glycoprotein, resistance to tamoxifen, and experimental pulmonary metastases ofthe R3230AC rat mammary adenocarcinoma in vivo (Kellen et al. 1991). Therefore,the potential for clinical enhancement of standard chemotherapeutic drugs medi-ated by verapamil and its derivatives remains on the horizon (Chatterjee et al. 1990;Mickisch et al. 1991a; Hollt et al. 1992; Kroemer et al. 1992; Teodori et al. 2005;Shen et al. 2008).
Calmodulin is an intracellular calcium-binding protein that plays critical roles in awide range of cellular activities (Ramakrishnan et al. 1989). Although the lack ofdown-regulation of calmodulin was found to produce higher levels of this proteinin transformed cells (Jaffr´ezou and Laurent 1993), such difference was not foundbetween the drug-sensitive and MDR P388 leukemia cells (Nair et al. 1986). How-ever, its calcium-sequestrating regulatory roles prompted investigation on the MDR-reversing effects of the potent calmodulin antagonist trifluoperazine (Tsuruo et al.
1982, 1983b; Klohs et al. 1986), and many antipsychotic phenothiazines marketedin the United States for clinical use and investigational compounds were found toreverse experimental MDR (Ganapathi et al. 1984; Ford et al. 1989; Ford et al. 1990;Fan et al. 1994b; Zhu et al. 2005).
2 Metastasis and Drug Resistance Antibiotics and Analogs
New drug development is time consuming and costly, hindering the availabilityof effective anticancer drug for the treatment of metastatic cancer. Similar to thecircumvention of clinical side effects of anticancer drugs (Tsuruo et al. 1983a, b),one approach to overcome drug resistance of cancer cells would be the developmentof derivatives amongst clinically proven chemotherapeutic compounds. Severalantibiotics such as the third-generation broad-spectrum cephalosporins (cefopera-zone and ceftriaxone) (Gosland et al. 1989), protein synthesis inhibitor antibacterialerythromycin (Hofsli and Nissen-Meyer 1989), veterinary antimicrobial monensin(Ling et al. 1995), a variety of vinca alkaloid derivatives (Ruiz et al. 1989; Nasioulaset al. 1990) were found to reverse experimental MDR phenotypes. Of particularinterest is the MDR-reversing effect of an anticancer benzylisoquinoline plant alka-loid thaliblastine that binds to P-glycoprotein and reverses doxorubicin resistanceof the P388 MDR cells (Chen et al. 1993). Its low toxicity (Todorov 1988) andstructural similarity to other compounds that have a photoaffinity for P-glycoprotein(Beck and Qian 1992) make it a potential chemosensitizer with a promising prospect(Pajeva et al. 2004).
Mdr-like genes exist across the entire phylogenetic spectrum. The reversal of MDRphenotypes in mammalian cells by calcium channel antagonists has functionalanalogies with the effects of agents circumventing chloroquine resistance in parasiteprotozoa (Bitonti et al. 1988), in which the drug-resistant phenotype was conferredby a protein coded by a gene closely related to mammalian mdr1 (Wilson et al.
1989). The indole-containing antimalarial quinine and structurally related com-pounds such as its anti-arrhythmic stereoisomer sodium channel blocker quinidinehave been found to produce an MDR-reversing activity (Tsuruo et al. 1984; Lehnertet al. 1991; Sato et al. 1991). Many of those compounds are neurohumoral antag-onists that include reserpine (antihypertensive and antipsychotic) and yohimbine(α-adrenergic blocker chemically similar to reserpine) (Fan et al. 1994a). Althoughit is clear that the functionality of the human mdr1 gene product is distinct from themalarial counterpart (Ginsburg and Krugliak 1992), the reversal of drug resistancein both systems by similar compounds implies the possibility of similar responsesfor action (Vezmar and Georges 2000).
Cyclosporins and Analogs
Cyclosporin A, the complex hydrophobic fungal cyclic undecapeptide, is commonlyused as an immunosuppressant for organ transplantation. At concentrations achiev-able clinically, it is considered one of the most effective MDR-reversing agents.
D. Fan et al.
The original reports of Slater et al. (1986a, b) initiated extensive studies on MDR-reversal, mediated by cyclosporins and related compounds (Twentyman et al. 1987;Chao et al. 1990; Dorr and Liddil 1991; Spoelstra et al. 1991; Loor et al. 1992;Arceci et al. 1992). Although cyclosporins have high affinity for P-glycoprotein(Goldberg et al. 1988; Foxwell et al. 1989), the reversal effects of cyclosporinsdid not correlate consistently with either drug accumulation (Slater et al. 1986a,b; Chambers et al. 1989; Hait et al. 1989) or direct interaction with P-glycoprotein(Hait et al. 1989). Nevertheless, cyclosporins and related compounds continue toproduce reversal of experimental MDR phenotypes (Shen et al. 2008). Its derivativeSDZ PSC-833 (Boesch et al. 1991; Ludwig et al. 2006; Shen et al. 2008) and thesemi-synthetic cyclic peptolide derivative SDZ 280–446 (Loor et al. 1992; Lehneet al. 2000) showed MDR-reversing effects superior even to those of cyclosporin A,which was about one order of magnitude more active than other known chemosen-sitizers such as verapamil.
Hormones and Antihormones
The induction, in pregnant murine uterus, of high levels of mdr1 mRNA, mediatedby estrogen and progesterone (Arceci et al. 1988), and the cross-resistance of MDRbreast carcinoma cells to antiestrogens with concomitant loss of estrogen recep-tors (Vickers et al. 1988) and progesterone receptors (Kacinski et al. 1989) initiatedextensive studies on the role of steroid hormones in MDR-reversal (Berman et al.
1991; Hu et al. 1991; Fleming et al. 1992; Stuart et al. 1992; Mutoh et al. 2006).
Although the effects of antiestrogens tamoxifen, toremifene, and 4-hydroxy tamox-ifen may be influenced by serum protein binding (Wurz et al. 1993; Chatterjee andHarris 1990), the reversal effects by the most active hormone progesterone havebeen shown to interact directly with P-glycoprotein (Yang et al. 1989; Naito et al.
1989; Safa et al. 1990). Subsequently, it was demonstrated that progesterone dis-tinguishes two mdr gene products (Yang et al. 1990) and specifically regulates theactivity of the mdr1b promoter via the A form of the progesterone receptor (Piekarzet al. 1993). However, results from clinical trials with vinblastine and high-dosemegestrol acetate were unremarkable (Matin et al. 2002).
Woodcock et al. (1990) found that Cremophor EL, a relatively inert formula ofpolyethoxylated castor oil commonly used as pharmaceutical emulsifier (e.g., forpreparations of water-insoluble compounds such as cyclosporins and taxol), couldreverse experimental MDR at attainable clinical concentrations. This reversal effecthas been since confirmed by using various MDR cells and pharmaceutical surfac-tants such as Solutol HS15 (Coon et al. 1991), Triton X-100, and Thesit (Fricheet al. 1990; Spoelstra et al. 1991; Woodcock et al. 1992). There was also evidence 2 Metastasis and Drug Resistance that nontoxic amounts of Cremophor EL and Tween 80 could effectively competefor P-glycoprotein binding with photoaffinity azidopine (Friche et al. 1990). Thereformulation of conventional anticancer agents to include sufficient but nontoxicconcentrations of these surfactants may enhance their clinical efficacy and over-come MDR.
A major limitation to the use of anticancer drugs is their nonspecific clinicaltoxicities that impair the therapeutic efficacy of these agents. Liposomes arebiodegradable, nonimmunogenic, and relatively nontoxic, and they can be safelyused to modify pharmacokinetic properties such as distribution, circulatory transittime, and drug metabolism, to target drugs and biologicals to most of the majororgans in animals and humans (Lopez-Berestein et al. 1984; Fogler et al. 1985), toavert systemic clinical toxicities (Forssen and Tokes 1981), and to improve therapeu-tic efficacy of antimicrobials (Lopez-Berestein et al. 1985), anticancer drugs (Huanget al. 1992; Ahmad et al. 1993), immunomodulators (Fidler 1988), and growth fac-tors (Schackert et al. 1989; Fan et al. 1989). Other studies have shown that liposomescomposed of various phospholipids can enhance the cytotoxicities of MDR-linkeddrugs such as doxorubicin, vinblastine, vincristine, and annamycin (Fan et al. 1990;Seid et al. 1991; Rahman et al. 1992; Thierry et al. 1993). Although the mode ofaction for MDR-reversal by liposomes is not clearly understood, the experimentalreversal of drug resistance by liposomes containing specific phospholipids has beenattributed to perturbation of the plasma membranes (Fan et al. 1990), to increasingdrug incorporation and intracellular redistribution (Thierry et al. 1993), or to directinteraction with P-glycoprotein (Thierry et al. 1993). The practical utility of lipo-some encapsulation in cancer treatment is obvious, but its mechanism remains to bedefined (Zalipsky et al. 2007).
The studies using calcium channel blockers and calmodulin antagonists to over-come MDR phenotypes continued. Dexniguldipine (B-859-35), the (–) isomer ofantihypertensive niguldipine, was found better than verapamil in reversing MDR(Hofmann et al. 1991; Reymann et al. 1993; Dietel et al. 1996; He and Liu 2002).
Various less toxic derivatives of verapamil (e.g., Ro11-2933) (Abderrabi et al. 1996),dihydropyridine (e.g., S16324, S16317) (Saponara et al. 2007), benzothiazepines(MDL 201,307) (Newman et al. 1996), and isoquinolinesulfonamides (e.g., W-77,CKA-1083) (Maeda et al. 1993), have been found potential agents for overcomingMDR. In the past years, many innovative molecules have also been investigated fortheir ability to circumvent MDR. Those findings included the studies of employ-ing MRK16 anti-P-glycoprotein antibody to reverse bone marrow drug resistance D. Fan et al.
of MDR transgenic mice (Mickisch et al. 1992a, b). Another anti-P-glycoproteinantibody, UIC2, was also effective in reversing experimental MDR (Mechetner andRoninson 1992). Other important compounds capable of reversing MDR includethose of the adenyl cyclase inhibitor forskolin (Wadler and Wiernik 1988; Morriset al. 1991; Yin et al. 2000), potassium-sparing diuretic amilorides (Epand et al.
1991; Miraglia et al. 2005), α- or β-adrenoceptor antagonists amiodarone (Lehn-ert et al. 1996) and SKB 105854 (Fan et al. 1994b), antidepressant trazodone (Fanet al. 1994a), antipsychotic benzquinamide (Mazzanti et al. 1992), triazine S 9788(Dhainaut et al. 1992; Moins et al. 2000), and the hydrophobic platelet anticoagulantdipyridamole (Verstuyft et al. 2003) and its derivative BIBW22BS (Schr¨oder et al.
1996). The antihistaminic terfenadine (Seldane) restored the sensitivity of MDRcells to doxorubicin (Hait et al. 1993). FB642 is a systemic benzimidazole fungi-cide with antitumor activity against a broad spectrum of tumors and drug-resistantand MDR cell lines (Hammond et al. 2001). Furthermore, it was found that introduc-tion of an MDR1-targeted small interfering RNA duplex into drug-resistant cancercells markedly inhibited the expression of MDR1 mRNA and P-gp and restoredsensitivity to multidrug-resistant cancer cells (Wu et al. 2003).
Clinical Reversal of MDR
Extensive clinical trials have been conducted in the past decade. As the first ofsuch reversing agents entering clinical trials, calcium channel blocker verapamilwas met with mixed outcomes that were discouraging in some studies (no response)(Rougier et al. 1969; Benson et al. 1985; Saltz et al. 1994) but were exception-ally promising in others (>50–70% response rate) (Cairo et al. 1989; Holmeset al. 1989; Figueredo et al. 1990; Miller et al. 1991; Salmon et al. 1991). Lym-phoma was consistently more responsive to the chemosensitizing effects of vera-pamil (Holmes et al. 1989; Miller et al. 1991; Chabner et al. 1994). Although thenumber of patients was small, the combination trial of chloroquine with conven-tional chemotherapy and radiotherapy was clinically effective in improving mid-term survival for glioblastoma multiforme (Sotelo et al. 2006). Unfortunately, thenumber of clinical studies of phenothiazines such as the calmodulin-inhibitor triflu-operazine (Miller et al. 1988; Murren et al. 1996) and antiemetic prochlorperazine(Sridhar et al. 1993; Raschko et al. 2000) was small, and the outcome was marginal.
Likewise, trials of doxorubicin derivative 4-iodo-4-deoxydoxorubicin (Sessa et al.
1992), antiestrogen tamoxifen (Stuart et al. 1992; Trump et al. 1992; Millwardet al. 1992), and calcium channel blocker nifedipine (Philip et al. 1992) were nothighly remarkable. However, isolated trials of the benzothiazepine calcium chan-nel blocker diltiazem and antimalarial quinine showed enhancement of leukemiaresponses to cytarabine, mitoxantrone, and vincristine (Bessho et al. 1985; Solaryet al. 1992). Although cyclosporin A may not be effective in enhancing the efficacyof epidoxorubicin in colon cancer patients (Verweij et al. 1992), a Southwest Oncol-ogy Group study showed responses in poor-risk acute myeloid leukemia patients 2 Metastasis and Drug Resistance receiving sequential treatments with cytarabine and daunomycin with concurrentinfusion of cyclosporin A (List et al. 1992). Although clinical correlations are want-ing (Holzmayer et al. 1992; Hait and Yang 2005), the use of biochemical modu-lation of clinical MDR remains a viable approach to improve treatments of cancerwith conventional chemotherapy (Fojo and Bates 2003).
Overview of Experimental MDR-Reversal
Many compounds, whose primary mechanism of action is blocking calcium chan-nels, have been found to have MDR-reversing activities. It is not clear why chem-icals that affect the flux of calcium in cells can elevate the accumulation ofMDR-associated natural chemotherapeutic drugs. Many MDR-reversing agents arecationic amphiphiles (contains both hydrophobic and polar regions) that are highlylysosomotropic (Jaffr´ezou et al. 1991; Akiyama et al. 1984; Ramakrishnan et al.
1989) and could modulate intracellular turnover and trafficking of specific phos-pholipids that may affect the MDR phenotype (Jaffr´ezou et al. 1992; Jaffr´ezouand Laurent 1993). These implications may be unorthodox because of the seemingdeparture from the functionality of P-glycoprotein. It must be noted that proteinsare rigid in nature and their functionality (mechanisms of action) and efficiency(kinetics) are often conformationally regulated by phospholipid matrix. This wasshown by restoration of calcium accumulation across lipid bilayers by reconstitutivesarcoplasmic reticulum phospholipid-mediated, calcium- and magnesium-activatedATPase activity (Racker 1972) and by alterations in lipid fluidity that modulatesP-glycoprotein-mediated drug transport in rat liver canalicular membrane vesicles(Sinicrope et al. 1992). Amplification of mdr1 and overexpression of its messen-ger underline that typical MDR phenotypes are probably by way of induction viaplasma membrane with specific chemotherapeutic drugs.
Unlike proteins, phospholipid compositions are remarkably different in trans- formed and tumor cells (Bergelson et al. 1970; Hatten et al. 1977), and thelipolytic activity of neoplasms is accentuated (Elwood and Morris 1968; Fran-son Patriaria and Elsbach 1974). The negative headgroup of the commonly inter-nal phosphatidylserine (externalized in undifferentiated and neoplastic cells) hashigh affinity for calcium via coordination-chelation bonds (Paphadjopoulous 1968)that could initiate a localized drop in pH and a lateral phase separation of phos-phatidylserine in the plasma membrane (Ohnishi and Ito 1973; Tr¨auble and Eibl1974). Acidic pH can produce a reversible nonbilayer inverted micelle type in mem-branes containing phosphatidylserine (Hope and Cullis 1980). In addition to theregulatory effects of calcium and pH, phosphatidylserine was found deacylated bya unique membrane-associated phospholipase-A in SV40-transformed 3T3 fibrob-lasts and in human gastric carcinoma cells (Fan and Voelz 1977, 1980, 1984) to gen-erate short-lived but fusogenic lysophosphatidylserine (Stein Y and Stein O 1966;Ahkong et al. 1973). Therefore, the unique properties of plasma membrane lipidssuch as phosphatidylserine allow them to participate in biological phenomena by D. Fan et al.
means of a transient rearrangement of the membrane structure similar to that seenin MDR P388 leukemic cells (Garcia-Segura et al. 1992), and it regulates the effi-ciency (conformational changes and kinetic enhancements) of the efflux pump. Ithas been demonstrated that MDR phenotype of the human KB-V1 cell line (Ambud-kar et al. 1992) and expression of the human mdr1 in Sf9 insect cells can generatea high-capacity drug-stimulated membrane ATPase (Sarkadi et al. 1992), and alter-ations in lipid fluidity can modulate P-glycoprotein-mediated drug transport in ratliver canalicular membrane vesicles (Sinicrope et al. 1992) and partially in the puri-fied MDR CHRC5 Chinese hamster ovary cell plasma membrane P-glycoprotein(Doige et al. 1993). Therefore, the ATP-dependent calcium channel P-glycoproteinin cardiac and tumor cells could be retarded by blocking agents via a "nonspecific"perturbation (evidenced by the manifold variety of effectors) of the plasma mem-brane phospholipids to conformationally hinder ATPase activity (energy supply)and consequentially the functions of calcium channel and P-glycoprotein.
Metastasis and Drug Resistance
The various modes of MDR-reversal put into effect that cancer is biologicallyheterogeneous and metastatic cells are the champions of survival. The process oftumor metastasis is highly selective and consists of a series of sequential and unifiedsteps (Fidler 1990). Despite improvements in diagnosis, surgical techniques, patientcare, and adjuvant therapies, most deaths from cancer are due to metastases that areresistant to conventional therapies. The major obstacle to effective treatment is thebiologic heterogeneity of tumor cells. Moreover, metastases can be located in lymphnodes and different organs, and the specific organ microenvironment influences thebiologic behavior of metastatic cells, including their response to systemic therapy(Fidler 2002). One of the factors is the development of drug resistance phenotype inmetastatic cancer cells (Dutour et al. 2007; La Porta 2007). While cancer metastasesare of clonal origin (Talmadge et al. 1982), variant clones with diverse phenotypescan form and rapidly result in the generation of significant cellular diversity withinindividual metastases (Fidler and Hart 1982). The outcome of the metastatic processdepends on multiple and complex interactions of the metastatic cells with the hosthomeostatic mechanisms (Liotta et al. 1991; Fidler 1997).
We determined whether the expression level of metastasis-related genes is reg- ulated by specific organ microenvironments. Highly metastatic clones of humanprostate cancer were implanted into the prostate (orthotopic site) and subcutis(ectopic site). Tumors were harvested and processed for in situ hybridization (ISH)analysis. Spontaneous metastases in the lymph nodes were also evaluated. Tumorsgrowing in the prostate exhibited higher levels of epidermal growth factor-receptor(EGF-R), basic fibroblast growth factor (bFGF), interleukin (IL)-8, type IV collage-nase, and the multidrug resistance (mdr-1) gene than those growing in the subcutis(Greene et al. 1997).
2 Metastasis and Drug Resistance The orthotopic implantation of human cancer cells was mandatory for analysis of metastasis-related genes. Specifically, highly metastatic cells expressed highermRNA levels of type IV collagenase (which affects invasion), bFGF and IL-8(which affect angiogenesis), and mdr-1 compared with cells of low metastaticpotential. No difference in EGF-R expression (which affects growth) was foundbetween the cells, but the expression of E-cadherin (which affects cell cohesion)was decreased in the metastatic cells. Vascular endothelial growth factor/vascularpermeability factor (VEGF/VPF), which affects tumor angiogenesis, has also beenfound to be overexpressed in prostate cancer in comparison with normal epithe-lium or benign prostatic hyperplasia. We found that VEGF/VPF levels correlatedwith microvessel density and metastatic potential of human prostate cancer cellsgrowing in the prostate of nude mice (Balbay et al. 1999). Furthermore, there isan intratumoral heterogeneity of expression of tyrosine kinase growth receptorsfound in human colon cancer surgical specimens and in orthotopic tumors (Kuwaiet al. 2008). Collectively, these data suggest that the expression level of metastasis-regulating genes by metastatic cells can be induced by factors in the organ microen-vironment and can influence the drug resistant phenotype of metastases.
Since the expression of various cytokines, growth factors, and their receptors on metastatic tumor cells and their microenvironment (e.g., endothelial cells) caninteract to provide survival advantage and mediate MDR phenotype, combiningchemotherapy and targeting the receptors of specific tyrosine protein kinases maybe effective in treating metastatic diseases. Our group has been successful in suchcombination therapies against several metastatic cancers in orthotopic animal mod-els. These included inhibiting the EGFR signaling by PKI166 in human renal cellcarcinoma growing orthotopically in nude mice (Kedar et al. 2002); targeting theexpression of platelet-derived growth factor receptor (PDGF-R) by STI571 (Kimet al. 2006); targeting EGF-R by PKI166 and VEGF-R by AEE788 with irinotecanin orthotopic colon carcinoma (Kitadai et al. 2006; Sasaki et al. 2007); simultane-ously inhibiting EGF-R/VEGF-R by AEE788 and PDGF-R by STI571 with gem-citabine against human pancreatic carcinoma (Yokoi et al. 2005); targeting tumorcells and tumor-associated endothelial cells in human prostate cancer cells growingin the bone of nude mice by inhibiting EGF-R using PKI166 (Kim et al. 2003);inhibiting EGF-R by PKI166 and PDGF-R by STI571 with taxol (Kim et al. 2004);or STI571 with zoledronate and paclitaxel (Kim et al. 2005).
The survival and growth of cells is dependent on an adequate supply of oxygenand nutrients and on the removal of toxic molecules. Oxygen can diffuse fromcapillaries for only 150–200 μm. When distances of cells from a blood supplyexceed this, cell death follows (Gimbrone et al. 1974; Folkman and Klagsbrun 1987;Kerbel and Folkman 2002; Fidler and Ellis 2004). Thus, the expansion of tumormasses beyond 1 mm in diameter depends on neovascularization, i.e., angiogenesis D. Fan et al.
(Folkman 1986). The formation of new vasculature consists of multiple, interdepen-dent steps. It begins with local degradation of the basement membrane surround-ing capillaries, followed by invasion of the surrounding stroma and migration ofendothelial cells in the direction of the angiogenic stimulus (Fidler et al. 2005).
Proliferation of endothelial cells occurs at the leading edge of the migrating col-umn, and the endothelial cells begin to organize into three-dimensional structuresto form new capillary tubes (Auerbach and Auerbach 1994). Differences in cellu-lar composition, vascular permeability, blood vessel stability, and growth regulationdistinguish vessels in neoplasms from those in normal tissue (Fidler and Ellis 1994).
The onset of angiogenesis involves a change in the local equilibrium between proangiogenic and antiangiogenic molecules (Fidler 2001; Kerbel and Folkman2002). Some of the common proangiogenic factors include bFGF, which inducesproliferation in a variety of cells and has also been shown to stimulate endothelialcells to migrate, to increase production of proteases, and to undergo morphogene-sis (Folkman and Klagsbrun 1987). Likewise, VEGF/VPF has been shown to inducethe proliferation of endothelial cells, to increase vascular permeability, and to induceproduction of urokinase plasminogen activator by endothelial cells (Dvorak 1986;Dvorak et al. 1995). Additional proangiogenic factors include IL-8, a cytokine pro-duced by a variety of tissues and blood cells (Singh et al. 1994; Yoneda et al. 1998),platelet-derived endothelial cell growth factor, which has been shown to stimulateendothelial cell DNA synthesis and to induce production of FGF (Kim et al. 2005,2006), hepatocyte growth factor (HGF), or scatter factor, that increases endothelialcell migration, invasion, and the production of proteases (Bussolino et al. 1992), andplatelet-derived growth factor (PDGF) (Risau et al. 1992).
Moreover, the structure and architecture of tumor vasculature can dramatically differ from those found in normal organs (Ebhard et al. 2000; Nor and Pulverini1999; Nels et al. 1992). Indeed, blood vessels in tumors are different than thosefound in wound healing, and inflamed tissues. The blood flow through tumors can betortuous and is characterized by regions of necrosis, rapid cell division, and presenceof infiltrate cells. Receptors for VEGF (KDR in humans, Flt-1 in mice) are expressedspecifically by tumor endothelium as well as the angiopoietin tyrosine kinase recep-tor, Tie-2 (reviewed in Liu et al. 2000). In addition, receptors for PDGF and EGFare found on tumor endothelial cells (Uehara et al. 2003; Suhardja and Hoffman2003). The endothelium is fragile, and upregulation of survival factors (such asBcl-2 and survivin) by molecules found in abundance within the tumor microen-vironment such as VEGF and bFGF helps to prevent apoptosis of new endothelium(Wang et al. 2002; Karsan et al. 1997; Gerber et al. 1998). There is increased leak-iness to macromolecules (perhaps due to the presence of VEGF) (Jain 1987; Dvo-rak 1990), and vessels often lose distinct features of arteriole, capillary, and venuleformation. Modern techniques, such as phage-display targeting, have defined "vas-cular addresses" that may be distinct for different organs as well as tumors in thoseorgans and perhaps offer attractive targets for antivasculature therapy (Pasqualiniet al. 2002).
Angiogenic heterogeneity exists within a single tumor (zonal or intralesional) between different metastases even in a single organ, and different neoplasms of the 2 Metastasis and Drug Resistance same histologic type are also documented (Kumar et al. 1998; Yu et al. 2001). Forexample, the expression of proangiogenic molecules (and, therefore, blood vesseldensity) in murine or human tumors growing at orthotopic sites in athymic miceis zonal, i.e., demonstrates intralesional heterogeneity. Small tumors (3–4 mm indiameters) expressed more bFGF and IL-8 than large tumors (>10 mm in diame-ters), whereas more VEGF is expressed in large tumors. Immunostaining showed aheterogeneous distribution of these angiogenic factors within the tumor; expressionof bFGF and Il-8 was highest on the periphery of a large tumor, where cell divi-sion was maximal. VEGF expression was higher in the center of the tumor (Kumaret al. 1998). Similarly, heterogeneous dependence on angiogenesis was reported forcell subpopulations isolated from human melanoma xenografts having differentialexpression of hypoxia-inducing factor-1 (Yu et al. 2001).
Heterogeneity of blood vessel distribution in surgical specimens of human cancers is well documented (Weidner et al. 1992). Benign neoplasms are spar-ely vascularized and tend to grow slowly in contrast to highly vascularized andrapidly growing malignant tumors (Weidner et al. 1992). The distribution of ves-sels in a tumor, however, is not uniform, and Weidner et al. cautioned that topredict the aggressive nature of human cancers, one must determine the mean ves-sel density (MVD) in the "areas of most intense neovascularization", i.e., tumorsexhibit intralesional and zonal heterogeneity for MVD (Weidner et al. 1992; Jain1987, 2008). Similarly, the expression of proangiogenic molecules in surgical speci-mens of human colon carcinoma was determined by in situ hybridization technique.
Matrix metalloproteinase-9 and bFGF were overexpressed at the periphery of thetumor where cells were rapidly dividing, whereas VEGF expression was higher inthe center of the lesions (Kitadai et al. 1995).
The extent of angiogenic heterogeneity in malignant neoplasms is also regu- lated by the organ microenvironment. For example, human renal carcinoma cellsimplanted into the kidney of athymic mice produced a high incidence of lungmetastasis, whereas those implanted subcutaneously did not (Singh et al. 1994).
Histopathologic examination of the tissues revealed that the tumors grown in thesubcutis of nude mice had few blood vessels, as compared to tumors in the kidney.
The subcutaneous tumors also had a significantly lower level of mRNA transcriptsfor bFGF than tumor in the kidney, and the expression of the naturally occurringangiogenic inhibitor, IFN-β (which downregulates bFGF) was high in epithelialcells and fibroblasts surrounding the subcutaneous tumors. This was not detected inor around tumors grown in the kidney (Singh et al. 1995). The production of IL-8 bymelanoma cells is regulated by complex interactions with skin keratinocytes (Her-lyn 1990). IL-8 expression can be increased by co-culture of melanoma cells withskin keratinocytes, and this expression is inhibited by coincubation of melanomacells with hepatocytes from the liver (Gutman et al. 1995). The organ microenviron-ment also influences the expression of VEGF. Human gastric cancer cells implantedinto the stomach were highly vascularized and expressed high levels of VEGF, ascompared to implantation into an ectopic (subcutaneous) site, such as the skin. Inaddition, metastasis only occurred from the tumor implanted in the stomach (Taka-hashi et al. 1996).
D. Fan et al.
The molecular cross-talk that occurs with tumor cells and endothelium within the tumor microenvironment results in sufficient recruitment of a vascular sup-ply that has physiological properties that allow migration and eventual escape ofsubpopulations of tumor cells able to complete a cascade of events necessary formetastasis.
Antivascular Therapy of MDR Prostate Carcinoma
Cancer of the prostate is the most common cancer affecting men in North Americaand is the second leading cause of cancer-related deaths. Mortality from prostatecancer usually results from the metastasis of hormone-refractory cancer cells.
Reports examining the pattern of metastasis in advanced prostate cancer indicatethat dissemination to bone and lymph nodes occurs in over 80% of the cases (Gar-nick and Fair 1996). The pathophysiology of prostate cancer bone metastases iscomplex and involves the interaction of tumor cells with osteoclasts, osteoblasts,endothelial cells and an assortment of regulatory proteins (e.g., steroid hormones,cytokines, and growth factors).
To study the factors that are critical for growth of prostate cancer cells in the bone, we established a murine model of hormone-refractory prostate cancer bonemetastasis. To generate prostate cancer growth in the bone, we performed a per-cutaneous intraosseal injection on nude mice by inserting a 27-gauge needle intothe tibia immediately proximal to the tuberositas tibia (Uehara et al. 2003). Afterpenetrating the cortical bone, we deposited 20 μl of tumor cell suspension (2 ×105 androgen-independent PC3-MM2 cells) in the bone cortex with the use of acalibrated, push button-controlled dispensing device. Five weeks later, we resectedthe tumor-bearing leg and performed an extensive immunohistochemical survey ofthe bone lesions in an effort to identify potential factors that may be involved inthe regulation of prostate tumor cell growth. A preliminary immunohistochemicalevaluation revealed robust tumor cell expression of bFGF, VEGF, IL-8, PDGF BB,and its receptor PDGFR-Rβ. Expression of these proteins was most pronounced intumors that were growing adjacent to the bone. In contrast, in those tumors thathad lyzed the bone and extended their growth to include the surrounding muscle,we detected only minimal levels of the angiogenic proteins, suggesting that factorswithin the bone environment were influencing the phenotype of the tumor cells.
A more comprehensive examination of distribution pattern of PDGFRβ revealed that PDGFR-β was present on both prostate tumor cells and on tumor-associatedendothelium and that, moreover, this receptor tyrosine kinase was activated. Phos-phorylated PDGFR-β was not found in either the contralateral nontumor leg or intumor cells growing away from the bone, i.e., in the muscle. These findings indicatethat the PDGF BB produced by tumor cells was acting in an autocrine manner tostimulate tumor cells and in a paracrine fashion to convey information to tumor-associated endothelial cells. The expression pattern of activated PDGFR-β in thebone metastases suggested that it might be a target for therapy in that inhibition of 2 Metastasis and Drug Resistance this signaling cascade could potentially affect both the malignant cell population andthe tumor blood supply. To test this hypothesis, we treated mice with experimentalbone metastasis using the tyrosine kinase inhibitor of PDGFR-β, STI571 (imatinibmesylate, Gleevec). In mice treated with STI571 or the combination of STI571 pluspaclitaxel, we found induction of significant apoptosis of endothelial cells and tumorcells that resulted in inhibition of tumor growth, a significant decrease of lymphaticmetastases, and a significant decrease of bone lysis (Uehara et al. 2003). Theseexperiments demonstrated that tumor-associated endothelial cells express phospho-rylated PDGFR when adjacent tumor cells express PDGF, and that inhibition of thisactivation with a PDGFR tyrosine kinase inhibitor, particularly in combination withchemotherapy, can produce significant therapy.
To determine the molecular mechanism for the antiangiogenic effects observed on the tumor-associated endothelial cells in vivo, we established cultures of murinebone microvascular endothelial cells and examined their response to stimulationwith PDGF BB ligand and to blockade of PDGFR signaling with STI571 (Lang-ley et al. 2004). Cultured bone endothelial cells expressed PDGFR-β, and PDGFBB-induced phosphorylation on these cells could be inhibited by STI571 in a dose-dependent manner. Stimulation of the bone endothelial cells with PDGF BB resultedin activation of Akt and ERK1/2, and this signaling cascade could be completelyabrogated by STI571. In addition, we found that bone endothelial cells respondto PDGF BB by increasing their cell division and upregulating the anti-apoptoticprotein Bcl-2. We then examined the response of bone endothelial cells to treat-ment with STI571 and taxol. Treatment of bone endothelial cells with only a singleagent produced little effect. However, the combined treatment of STI571 and taxolresulted in a significant increase in the number of cells expressing activated caspase-3 and a concomitant decline in Bcl-2. Consistent with these results, we found thatwhen bone endothelial cells were confronted with both STI571 and low levels oftaxol, there was a threefold increase in their cytotoxicity.
Collectively, these data suggest that a primary target for the STI571 and pacli- taxel therapy may be the tumor-associated blood vessels. To test this hypothesis,we established a multidrug resistant prostate cell line by chronically exposing PC3-MM2 cells to increasing concentrations of taxol (Kim et al. 2006). The resultingcell line, PC3-MM2-MDR, is 70 times more resistant to paclitaxel in vitro, and thegrowth of the cells is not affected by treatment with paclitaxel or the combinationof paclitaxel and STI571. When the PC3-MM2-MDR cells were implanted into thebone microenvironment, they displayed the same angiogenic profile as the parentalcell line. Endothelial cells in normal tissues rarely divide, whereas 2–3% of theendothelial cells in prostate cancer divide daily (Augustin et al. 2002; Eberhard et al.
2000). These dividing endothelial cells should be sensitive to anticycling drugs suchas paclitaxel. Nevertheless, in the present experiment, paclitaxel did not decreasethe MVD appreciably, likely because of the fact that stimulation of endothelial cellswith PDGF leads to resistance to paclitaxel, and that blockade of PDGF-R phospho-rylation with imatinib reverses the resistance to paclitaxel (Langley et al. 2003). Asstated above, the first wave of apoptosis in bone tumors from mice treated with ima-tinib and paclitaxel for only 2 weeks occurred in tumor-associated endothelial cells, D. Fan et al.
followed by apoptosis of tumor cells and ultimately tumor necrosis. By the fourthweek of treatment with imatinib and paclitaxel or imatinib alone, concurrent apop-tosis of tumor cells and tumor-associated endothelial cells was observed. Withoutpaclitaxel, imatinib may produce therapeutic effects by the blockade of PDGF-R,which serves as a survival factor (Langley et al. 2003).
Thus, the imatinib-induced blockade of PDGF-R combined with paclitaxel appears to target the tumor-associated endothelial cells. Whether this approach canbe useful for other types of tumors is unknown. The heterogeneity of angiogenesisin human tumors and the findings that endothelial cells of different organs are phe-notypically distinct (Langley and Fidler 2007) indicate that further investigations tounderstand the interaction of different types of tumor cells and endothelial cells indifferent organs are necessary for the development of optimal regimens of targetedantivascular therapies. For this reason, we performed another series of experimentsusing the multidrug resistant PC-3MM2-MDR cells growing in the prostate of nudemice and treated the mice with paclitaxel and the tyrosine kinase inhibitor, AEE788,that targets phosphorylation of EGF-R/VEGF-R (Busby et al. 2006). The significantinhibition of local tumor growth and lymph node metastases again demonstratedthat tumor-associated endothelial cells, rather than the tumor cells, were the pri-mary target of the chemotherapy. Those studies provide a better understanding ofthe molecular mechanisms that regulate the process of metastasis and of the com-plex interactions between the metastatic cells and the organ microenvironment (Kimet al. in press).
Abderrabi, M., Marchal, S., and Merlin, J. L. 1996. Comparative in vitro evaluation of dithiane analogs of tiapamil, Ro 11-2933, Ro 44-5911 and Ro 44-5912 as multidrug resistance modula-tors. Anticancer Drugs 7:430–436.
Adachi, T., Nakagawa, H., Chung, I., Hagiya, Y., Hoshijima, K., Noguchi, N., Kuo, M. T., and Ishikawa, T. 2007. Nrf2-dependent and -independent induction of ABC transporters ABCC1,ABCC2, and ABCG2 in HepG2 cells under oxidative stress. J. Exp. Ther. Oncol. 6:335–348.
Aftab DT, Yang JM, Hait WN. 1994. Functional role of phosphorylation of the multidrug trans- porter (P-glycoprotein) by protein kinase C in multidrug-resistant MCF-7 cells. Oncol. Res.
6:59–70.
Ahkong, Q. F., Cramp, F. C., Fisher, D., Howell, J. I., Tampion, W., Verrinder, M., and Lucy, J.
A. 1973. Chemically-induced and thermally-induced cell fusion:lipid-lipid interactions. Nature(New Biol.) 242:215–217.
Ahmad, I., Longenecker, M., Samuel, J., and Allen, T. M. 1993. Antibody-targeted delivery of doxorubicin entrapped in sterically stabilized liposomes can eradicate lung cancer in mice.
Cancer Res. 53:1484–1488.
Akiyama, S., Cornwell, M. M., Kuwano, M., Pastan, I., and Gottesman, M. M. 1988. Most drugs that reverse multidrug resistance inhibits photoaffinity labeling of P-glycoprotein by a vinblas-tine analog. Mol. Pharmacol. 33:144–147.
Akiyama, S., Gottesman, M. M., Hanover, J. A., FitzGerald, D. J. P., Willingham, M. C., and Pastan, I. 1984. Verapamil enhances the toxicity of conjugates of epidermal growth factorwith Pseudomonas exotoxin and antitransferrin receptor with Pseudomonas exotoxin. J. CellPhysiol. 120:271.
2 Metastasis and Drug Resistance Alley, M. C., Scudiero, D. A., Monks, A., Hursey, M. L., Czerwinski, M. J., Fine, D. L., Abbott, B. J., Mayo, J. G., Shoemaker, R. H., and Boyd, M. R. 1988. Feasibility of drug screening with panelsof human tumor cell lines using a microculture tetrazolium assay. Cancer Res. 48:589–601.
Ambudkar, S. V., Lelong, I. H., Zhang, J., Cardarelli, C. O., Gottesman, M. M., and Pastan, I. 1992.
Partial purification and reconstitution of the human multidrug-resistance pump: characteriza-tion of the drug-stimulatable ATP hydrolysis. Proc. Natl. Acad. Sci. USA 89:8472–8476.
Arceci, R. J., Croop, J. M., Horwitz, S. B., and Housman, D. 1988. The gene encoding multidrug resistance is induced and expressed at high levels during pregnancy in the secretory epitheliumof the uterus. Proc. Natl. Acad. Sci. USA 85:4350–4354.
Arceci, R. J., Stieglitz, K., and Bierer, B. E. 1992. Immunosuppressants FK506 and rapamycin function as reversal agents of the multidrug resistance phenotype. Blood 80:1528–1536.
Auerbach, W., and Auerbach, R. 1994. Angiogenesis inhibition: a review. Pharmacol. Ther.
Augustin, H. G., Kmeta, J., Alves, F., Baumbach, J., Eberhard, A., Kahlert, S., and Dandekar, G.
2002. Quantitating angiogenesis and assessing the causal relationship between angiogenesisand tumorigenesis: problems and progress. Ann. Hematol. 81(suppl. 2):68–72.
Balbay, D., Pettaway, C. A., Kuniyasu, H., Inoue, K., Ramirez, E., Li, E., and Fidler, I. J. 1999.
Highly metastatic human prostate cancer growing within the prostate of athymic mice overex-presses a vascular endothelial growth factor. Clin. Cancer Res. 5:783–789.
Beck, W. T., Cirtain, M. C., Glover, C. J., Felsted, R. L., and Safa, A. R. 1988. Effects of indole alkaloids on multidrug resistance and labeling of P-glycoprotein by a photoaffinity analog ofvinblastine. Biochem. Biophys. Res. Commun. 153:959–966.
Beck, W. T., and Qian, X. D. 1992. Photoaffinity substrates for P-glycoprotein. Biochem. Pharma- col. 43:89–93.
Benson, A. B., Trump, D. L., Koeller, J. M., Egorin, M. I., Olman, E. A., Witte, R. S., Davis, T.
E., and Tormey, D. C. 1985. Phase I study of vinblastine and verapamil given by concurrent ivinfusion. Cancer Treat. Rep. 69:795–799.
Bergelson, L. D., Dyatlovitskaya, E. V., Torkhovskaya, T. I., Sorokina, I. B., and Gorkova, N.
P. 1970. Phospholipid composition of membranes in the tumor cell, Biochim. Biophys. Acta,210:287.
Berman, E., Adams, M., Duigou-Osterndorf, R., Godfrey, L., Clarkson, B., and Andreeff, M.
1991. Effect of tamoxifen on cell lines displaying the multidrug-resistant phenotype. Blood 78:1385–1387.
Bessho, F., Kinumaki, H., Kobayashi, M., Habu, H., Nakamura, K., Yokota, S., Tsuruo, T., and Kobayashi, N. 1985. Treatment of children with refractory acute lymphocytic leukemia withvincristine and diltiazem. Med. Pediatr. Oncol. 13:199–202.
Bitonti, A. J., Sjoerdsma, A., McCann, P. P., Kyle, D. E., Oduola, A. M. J., Rossan, R. N., Milhous, W. K., and Davidson, D. E. Jr. 1988. Reversal of chloroquine resistance in malaria parasiteplasmodium falciparum by desipramine. Science 242:1301–1303.
Boesch, D., Muller, K., Pourtier-Manzanedo, A., and Loor, F. 1991. Restoration of daunomycin retention in multidrug-resistant P388 cells by submicromolar concentrations of SDZ PSC 833,a nonimmunosuppressive cyclosporin derivative. Exp. Cell Res. 196:26–32.
Bucana, C. D., Giavazzi, R., Nayar, R., OBrian, C. A., Seid, C., and Fan, D. 1990. Retention of vital dyes correlates inversely with the multidrug resistant phenotype of Adriamycin-selectedmurine fibrosarcoma variants. Exp. Cell Res. 190:69–75.
Busby, J. E., Kim, S. J., Yazici, S., Nakamura, T., Kim, J. S., He, J., Maya, M., Wang, X., Do, K.
A., Fan, D., and Fidler, I. J. 2006. Therapy of multidrug resistant prostate cancer by targetingthe epidermal growth factor receptor and vascular endothelial growth factor receptor on tumor-associated endothelial cells. Prostate 66:1788–1798.
Bussolino, F., DiRenzo, M. F., Ziche, M., Bocchietto, E., Olivero, M., Naldini, L., Gaudino, G., Tamagnone, L., Coffer, A., and Comoglio, P. M. 1992. Hepatocyte growth factor is a potentangiogenic factor which stimulates endothelial cell motility and growth. J. Cell Biol. 119:629–641.
D. Fan et al.
Cairo, M. S., Siegel, S., Anas, N., and Sender, L. 1989. Clinical trial of continuous infusion ver- apamil, bolus vinblastine, and continuous infusion VP-16 in drug-resistant pediatric tumors.
Cancer Res. 49:1063–1066.
Chabner, B. A., Bates, S. E., Fojo, A. T., Spolyar, M., and Wilson, W. H. 1994. Drug resistance in adult lymphomas. Semin. Hematol. 31:70–87.
Chambers, S. K., Hait, W. N., Kacinski, B. M., Keyes, S. R., and Hand-Schumacher, R. E. 1989. Enhancement of anthracycline growth inhibition in parent and multidrug-resistant Chinese hamster ovary cells by cyclosporin A and its analogues. Cancer Res. 49:6275–6279.
Chao, N. J., Aihara, M., Blume, K. G., and Sikic, B. I. 1990. Modulation of etoposide (VP-16) cytotoxicity by verapamil or cyclosporine in multidrug-resistant human leukemic cell lines andnormal bone marrow. Exp. Hematol. 18:1193–1198.
Chatterjee, M., and Harris, A. L. 1990. Reversal of acquired resistance to Adriamycin in CHO cells by tamoxifen and 4-hydroxy tamoxifen:role of drug interaction with alpha 1 acid glycoprotein.
Br. J. Cancer 62:712–717.
Chatterjee, M., Robson, C. N., and Harris, A. L. 1990. Reversal of multidrug resistance by vera- pamil and modulation by alpha 1-acid glycoprotein in wild-type and multidrug-resistant Chi-nese hamster ovary cell lines. Cancer Res. 50:2818–2822.
Chen, G., Ramachandran, C., and Krishan, A. 1993. Thaliblastine, a plant alkaloid, circumvents multidrug resistance by direct binding to P-glycoprotein. Cancer Res. 53:2544–2547.
Coon, J. S., Knudson, W., Clodfelter, K., Lu, B., and Weinstein, R. S. 1991. Solutol HS 15, nontoxic polyoxyethylene esters of 12-hydroxystearic acid, reverses multidrug resistance. Cancer Res.
51:897–902.
Cornwell, M. M., Pastan, I., and Gottesman, M. M. 1987. Certain calcium channel blockers bind specifically to multidrug resistant human KB carcinoma membrane vesicles and inhibit drugbinding to P-glycoprotein. J. Biol. Chem. 262:2166–2170.
Deffie, A. M., Alam, T., Seneviratne, C., Beenken, S. W., Batra, J. K., Shea, T. C., Henner, W.
D., and Goldenberg, G. J. 1988. Multifactorial resistance to Adriamycin:relationship of DNArepair, glutathione transferase activity, drug efflux, and P-glycoprotein in cloned cell lines ofAdriamycin-sensitive and -resistant P388 leukemia. Cancer Res. 48:3595–3602.
DeVita, V. T., Oliverio, V. T., Muggia, F. M., Wiernik, P. W., Ziegler, J., Goldin, A., Rubin, D., Henney, J., and Schepartz, S. 1979. The drug development and clinical trials programs of theDivision of Cancer Treatment, National Cancer Institute. Cancer Clin. Trials 2:195–216.
Dhainaut, A., Regnier, G., Atassi, G., Pierre, A., Leonce, S., Kraus-Berthier, L., and Prost, J. F.
1992., New triazine derivatives as potent modulators of multidrug resistance. J. Med. Chem.
35:2481–2496.
Dietel, M., Boss, H., Reymann, A., Pest, S., and Seidel, A. 1996. In vivo reversibility of mul- tidrug resistance by the MDR-modulator dexniguldipine (niguldipine derivative B859-35) andby verapamil. J. Exp. Ther. Oncol. 1:23–29.
Doige, C. A., Yu, X., and Sharom, F. J., 1993. The effects of lipids and detergents on ATPase-active P-glycoprotein. Biochim. Biophys. Acta 1146:65–72.
Dong, Z., Radinsky, R., Fan, D., Tsan, R., Bucana, C. D., Wilmanns, C., and Fidler, I. J. 1994.
Organ-specific modulation of steady-state mdr gene expression and drug resistance in murinecolon cancer cells. J. Natl. Cancer Inst. 86:913–920.
Dorr, R. T., and Liddil, J. D. 1991. Modulation of mitomycin C-induced multidrug resistance in vitro. Cancer Chemother. Pharmacol. 27:290–294.
Dutour, A., Leclers, D., Monteil, J., Paraf, F., Charissoux, J. L., Rousseau, R., and Rigaud, M. 2007.
Non-invasive imaging correlates with histological and molecular characteristics of an osteosar-coma model: application for early detection and follow-up of MDR phenotype. Anticancer Res.
27:4171–4178.
Dvorak, H. F., Brown, L. F., Detmar, M. and Dvorak, A. M. 1995. Vascular permeability fac- tor/vascular endothelial growth factor, Microvascular hyperpermeability, and angiogenesis.
Am. J. Pathol. 146:1029–1039.
2 Metastasis and Drug Resistance Dvorak, H. F. 1990. Leaky tumor vessels: consequences for tumor stroma generation and for solid tumor therapy. Prog. Clin. Biol. Res. 354A:317–330.
Dvorak, H. F. 1986. Tumors: wounds that do not heal. Similarities between tumor stroma genera- tion and wound healing. N. Engl. J. Med. 15(26):1650–1659.
Eberhard, A., Kahlert, S., Goede, V., Hemmerlein, B., Plate, K. H., and Augustin, H. G. 2000.
Heterogeneity of angiogenesis and blood vessel maturation in human tumors:implications forantiangiogenic tumor therapies. Cancer Res. 60:1388–1393.
Elwood, J. C., and Morris, H. P. 1968. Lack of adaptation in lipogenesis by hepatoma 9121, J.
Lipid Res. 9:337.
Epand, R. F., Epand, R. M., Gupta, R. S., and Cragoe, E. J., Jr. 1991. Reversal of intrinsic multidrug resistance in Chinese hamster ovary cells by amiloride analogs. Br. J. Cancer 63:247–251.
Fan, D., Baker, F. L., Khokhar, A. R., Ajani, J. A., Tomasovic, B., Newman, R. A., Brock, W.
A., Tueni, E., and Spitzer, G. 1988. Antitumor activity against human tumor samples of cis-diamminedichloroplatinum(II) and analogues at equivalent in vitro myelotoxic concentrations.
Cancer Res. 48:3135–3139.
Fan, D., Bucana, C. D., OBrian, C. A., Zwelling, L. A., Seid, C., and Fidler, I. J. 1990.
Enhancement of tumor cell sensitivity to Adriamycin by presentation of the drug inphosphatidylcholine-phosphatidylserine liposomes. Cancer Res. 50:3619–3626.
Fan, D., Fidler, I.J., Ward, N.E., Seid, C., Earnest, L.E., Housey, G.M., and OBrian, C.A. 1992a.
Stable expression of a cDNA encoding rat brain protein kinase C-ßI confers a multidrug-resistant phenotype on rat fibroblasts. Anticancer Res. 12:661–668.
Fan, D., Morgan, L. R., Schneider, C., Blank, H., and Fan, S. 1985. Cooperative evaluation of human tumor chemosensitivity in the soft-agar assay and its clinical correlations. J. CancerRes. Clin. Oncol. 109:23–28.
Fan, D., Poste, G., OBrian, C. A., Seid, C., Ward, N. E., Earnest, L. E., and Fidler, I. J. 1992b.
Chemosensitization of murine fibrosarcoma cells to drugs affected by the multidrug resistancephenotype by the antidepressant trazodone: an experimental model for the reversal of intrinsicdrug resistance. Int. J. Oncol. 1:735–742.
Fan, D., Poste, G., Ruffolo, R. R., Jr., Dong, Z., Seid, C., Earnest, L. E, Campbell, T. E., Clyne, R. K., Beltran, P. J., and Fidler, I. J. 1994a. Circumvention of multidrug resistancein murine fibrosarcoma and colon carcinoma cells by treatment with the α-adrenoceptor antag-onist furobenzazepine. Int. J. Oncol. 4:789–798.
Fan, D., Poste, G., Seid, C., Earnest, L. E., Bull, T., Clyne, R. K., and Fidler, I. J. 1994b. Reversal of multidrug resistance in murine fibrosarcoma cells by thioxanthene flupentixol. Invest. NewDrug 12:185–195.
Fan, D., Price, J., Schackert, H., Seid, C., Wilmanns, C., Chakrabarty, S., and Fidler, I. J. 1989.
Antiproliferative activity of liposome-encapsulated transforming growth factor-beta againstMDA-MB-435 human breast carcinoma cells. Cancer Commun. 1:337–343.
Fan, D., and Voelz, H. 1980. A plasma membrane-associated phospholipase in SV40-transformed 3T3 cells. Exp. Cell Res. 126:47–55.
Fan, D., and Voelz, H. 1984. Phospholipase activity in human tumors: localization by cytochemical staining. J. Cancer Res. Clin. Oncol. 107:242–244.
Fan, D., and Voelz, H. 1977. Phospholipid degradation by SV40-transformed murine fibroblasts.
Exp. Cell Res. 106:79–87.
Fidler, I. J. 2001. Angiogenic heterogeneity: regulation of neoplastic angiogenesis by the organ microenvironment (Editorial). J. Natl. Cancer Inst. 93:1040–1041.
Fidler, I. J. 1990. Critical factors in the biology of human cancer metastasis:twenty-eighth G.H.A.
Clowes memorial award lecture. Cancer Res. 50:6130–6138.
Fidler, I. J. 1995. Modulation of the organ microenvironment for treatment of cancer metastasis (Editorial). J. Natl. Cancer Inst. 84:1588–1592.
Fidler, I. J. 1997. Molecular biology of cancer: invasion and metastasis. In Cancer: Principles and Practice of Oncology, eds. V. T. DeVita, S. Hellman, and S. A. Rosenberg,pp 135–152.
Philadelphia: Lippincott-Raven.
D. Fan et al.
Fidler, I. J. 2001. Seed and soil revisited. Cancer Metastasis: Biol. Clin. Aspects 10:257–269.
Fidler, I. J. 1973. Selection of successive tumor lines for metastasis. Nature (New Biol.) 242: Fidler, I. J. 1988. Targeting of immunomodulators to mononuclear phagocytes for therapy of can- cer. Adv. Drug Deliv. Rev. 3:405–418.
Fidler, I. J. 2002. The organ microenvironment and cancer metastasis. Differentiation 70: Fidler, I. J. 2003. The pathogenesis of cancer metastasis: the ‘seed and soil' hypothesis revisited (Timeline). Nat. Rev. Cancer 3:3453–458.
Fidler, I. J. 1978. Tumor heterogeneity and the biology of cancer invasion and metastasis. Cancer Fidler, I. J. and Ellis, L. M. 2004. Neoplastic angiogenesis: All blood vessels are not created equal.
N. Engl. J. Med. (Perspective), 351:215–216.
Fidler, I. J., and Ellis, L. M. 1994. The implications of angiogenesis to the biology and therapy of cancer metastasis (Minireview). Cell, 79:185–188.
Fidler, I. J., and Hart, I. R. 1982. Biological diversity in metastatic neoplasms: origins and impli- cations. Science 217:998–1003.
Fidler, I. J., and Kripke, M. L. 1977. Metastasis results from preexisting variant cells within a malignant tumor. Science 197:893–895.
Fidler, I. J., Langley, R. R., Kerbel, R. S., and Ellis, L. M. 2005. Biology of cancer: angiogenesis.
In: Cancer: Principles and Practice of Oncology, 7th edt., eds. V. T, DeVita, Jr., S. Hellman, andS. A. Rosenberg,pp. 129–137. Philadelphia, PA: Lippincott & Wilkins.
Fidler, I. J., and Poste, G. 1985. The cellular heterogeneity of malignant neoplasms: implications for adjuvant chemotherapy. Semin. Oncol. 12:207–221.
Fidler, I. J., and Talmadge, J. 1986. Evidence that intravenously derived murine pulmonary melanoma metastases can originate from the expansion of a single tumor cell. Cancer Res.
46:5167–5171.
Figueredo, A., Arnold, A., Goodyear, M., Findlay, B., Neville, A., Normandeau, R., and Jones, A. 1990. Addition of verapamil and tamoxifen to the initial chemotherapy of small cell lungcancer. A phase I/II study. Cancer 65:1895–1902.
Fine, R. L., Koizumi, S., Curt, G. A., and Chabner, B. A. 1987. Effects of calcium channel blockers on human CFU-GM with cytotoxic drugs. J. Clin. Oncol. 5:489–495.
Fleming, G. F., Amato, J. M., Agresti, M., and Safa, A. R. 1992. Megestrol acetate reverses multidrug resistance and interacts with P-glycoprotein. Cancer Chemother. Pharmacol. 29:445–449.
Fogler, W. E., Wade, R., Brundish, D. E., and Fidler, I. J. 1985. Distribution and fate of free and liposome-encapsulated [3H]nor-muramyl dipeptide and [3H]muramyl tripeptide phos-phatidylethanolamine in mice. J. Immunol. 135:1372–1377.
Fojo, A. T., Akiyama, S., Gottesman, M. M., and Pastan, I. 1985. Reduced drug accumulation in multiply drug-resistant human KB carcinoma cell lines. Cancer Res. 45:3002–3007.
Fojo, A. T., and Bates, S. 2003. Strategies for reversing drug resistance. Oncogene 22: Folkman, J. 1986. How is blood vessel growth regulated in normal and neoplastic tissue? – Twenty- sixth GHA Clowes Memorial Award Lecture. Cancer Res. 46:467–473.
Folkman, J. and Klagsbrun, M. 1987. Angiogenic factors. Science 235:444–447.
Ford, J. M., Bruggeman, E. P., Pastan, I., Gottesman, M. M., and Hait, W. N. 1990. Cellular and biochemical characterization of thioxanthenes for reversal of multidrug resistance in humanand murine cell lines. Cancer Res. 50:1748–1756.
Ford, J. M., and Hait, W. N. 1990. Pharmacology of drugs that alter multidrug resistance in cancer.
Pharmcol. Rev. 42:155–199.
Ford, J. M., Prozialeck, W. C., and Hait, W. N. 1989. Structural features determining activity of phenothiazines and related drugs for inhibition of cell growth and reversal of multidrug resis-tance. Mol. Pharmacol. 35:105–115.
2 Metastasis and Drug Resistance Forssen, E. A., and Tokes, Z. A. 1981. Use of anionic liposomes for the reduction of chronic doxorubicin-induced cardiotoxicity. Proc. Natl. Acad. Sci. USA 78:1873–1877.
Foxwell, B. M. J., Mackie. A., Ling, V., and Ryffel, B. 1989. Identification of the mul- tidrug resistance-related P-glycoprotein as a cyclosporin binding protein. Mol. Pharmacol. 36:543–546.
Franson, R., Patriaria, P., and Elsbach, P. 1974. Phospholipid metabolism by phagocytic cells.
Phospholipase A2 associated with rabbit polymorphonuclear leukocyte granules. J. Lipid Res.
15:380–388.
Frei, E. 1982. The national cancer chemotherapy program. Science 217:600–606.
Friche, E., Jensen, P. B., Sehested, M., Demant, E. J., and Nissen, N. N. 1990. The solvents cre- mophor EL and Tween 80 modulate daunorubicin resistance in the multidrug resistant Ehrlichascites tumor. Cancer Commun. 2:297–303.
Ganapathi, R., Grabowski, D., Turinic, R., and Valenzuela, R. 1984. Correlation between potency of calmodulin inhibitors and effects on cellular levels and cytotoxic activity of doxorubicin(Adriamycin) in resistant P388 mouse leukemia cells. Eur. J. Cancer Clin. Oncol. 20:799–806.
Garcia-Segura, L. M., Soto, F., Planells-Cases, R., Gonzalez-Ros, J. M., and Ferragut, J. A. 1992.
Verapamil reverses the ultrastructural alterations in the plasma membrane induced by drugresistance. FEBS Lett. 314:404–408.
Garnick, M. B., and Fair, W. R. 1996. Prostate cancer:emerging concepts (Part II). Ann. Intern.
Med. 125:205–212.
Gerber, H. P., Dixit, V., and Ferrara, N. 1998. Vascular endothelial growth factor induces expres- sion of the anti-apoptotic proteins Bcl-2 and A1 in vascular endothelial cells. Am. J. Pathol.
158:1757–1765.
Gimbrone, M., Cotran, R. and Folkman, J. 1974. Tumor growth and neovascularization:an experi- mental model using rabbit cornea. J. Natl. Cancer Inst. 52:413–427.
Ginsburg, H., and Krugliak, M. 1992. Quinoline-containing anti-malarials–mode of action, drug resistance and its reversal. An update with unresolved puzzles. Biochem. Pharmacol. 43:63–70.
Goldberg, H., Ling, V., Wong, P. Y., and Skorecki, K. 1988. Reduced cyclosporin accumulation in multidrug-resistant cells. Biochem. Biophys. Res. Commun. 152:552–558.
Gosland, M. P., Lum, B. L., and Sikic, B. I. 1989. Reversal by cefoperazone of resistance to etopo- side, doxorubicin, and vinblastine in multidrug resistant human sarcoma cells. Cancer Res.
49:6901–6905.
Greene, G., Kitadai, Y., Pettaway, C. A., von Eschenbach, A. C., and Fidler, I. J. 1997. Corre- lation of metastasis-related gene expression with metastatic potential in prostate carcinomacells implanted in nude mice using an in situ mRNA hybridization technique. Am. J. Pathol.
150:1571–1582.
Gutman, M., Singh, R. K., Xie, K., Bucana, C. D., and Fidler, I. J. 1995. Regulation of IL-8 expression in human melanoma cells by the organ environment. Cancer Res. 55:2470–2475.
Hait, W. N., Gesmonde, J. F., Murren, J. R., Yang, J. M., Chen, H. X., and Reiss, M. 1993. Terfena- dine (Seldane):a new drug for restoring sensitivity to multidrug resistant cancer cells. Biochem.
Pharmacol. 26;45:401–406.
Hait, W. N., Stein, J. M., Koletsky, A. J., Harding. M. W., and Handschumacher, R. E. 1989.
Activity of cyclosporin A and a non-immunosuppressive cyclosporin on multidrug resistantleukemic cell lines. Cancer Commun. 1:35–43.
Hait, W. N., and Yang, J. M. 2005. Clinical management of recurrent breast cancer:development of multidrug resistance (MDR) and strategies to circumvent it. Semin. Oncol. 32:S16–S21.
Hamada, H., Okochi, E., Oh-hara, T., and Tsuruo, T. 1988. Purification of the Mr 22,000 calcium- binding protein (sorcin) associated with multidrug resistance and its detection with monoclonalantibodies. Cancer Res. 48:3173–3178.
Hammond, L. A., Davidson, K., Lawrence, R., Camden, J. B., Von Hoff, D. D., Weitman, S., and Izbicka, E. 2001. Exploring the mechanisms of action of FB642 at the cellular level. J. CancerRes. Clin. Oncol. 127:301–313.
D. Fan et al.
Harker, W. G., Bauer, D., Etiz, B. B., Newman, R. A., and Sikic, B. I. 1986. Verapamil-mediated sensitization of doxorubicin-selected pleiotrophic resistance in human sarcoma cells: selectivityfor drugs which produce DNA scission. Cancer Res. 46:2369–2373.
Hatten, M. E., Horwitz, A. F., and Burger, M. M. 1977. The influence of membrane lipids on the proliferation of transsformed and untransformed cell lines. Exp. Cell Res. 107:31.
He, L., and Liu, G. Q. 2002. Effects of various principles from Chinese herbal medicine on rhodamine123 accumulation in brain capillary endothelial cells. Acta Pharmacol Sin. 23:591–596.
Herlyn, M. 1990. Human melanoma: development and progression. Cancer Metastasis Rev. 9: Herzog, C. E., Tsokos, M., Bates, S. E., and Fojo, A. T. 1993. Increased mdr-1/P-glycoprotein expression after treatment of human colon carcinoma cells with P-glycoprotein antagonists. J.
Biol. Chem. 268:2946–2952.
Hofmann, J., Ueberall, F., Egle, A., and Grunicke, H. 1991. B-859–35, a new drug with anti-tumor activity reverses multi-drug resistance. Int. J. Cancer 47:870–874.
Hofsli, E., and Nissen-Meyer, J. 1989. Effect of erythromycin and tumour necrosis factor on the drug resistance of multidrug-resistant cells: reversal of drug resistance by erythromycin. Int. J.
Cancer 43:520–525.
Hollt, V., Kouba, M., Dietel, M., and Vogt, G. 1992. Stereoisomers of calcium antagonists which differ markedly in their potencies as calcium blockers are equally effective in modulating drugtransport by P-glycoprotein. Biochem. Pharmacol. 43:2601–2608.
Holmes, J., Jacobs, A., Carter, G., Janowska-Wieczorek, A., and Padua, R. A. 1989. Multidrug resistance in haematopoietic cell lines, myelodysplastic syndromes and acute myeloblasticleukemia. Br. J. Haematol. 72:40–44.
Holzmayer, T. A., Hilsenbeck, S., Von Hoff, D. D., and Roninson, I. B. 1992. Clinical correlates of MDR1 (P-glycoprotein) gene expression in ovarian and small-cell lung carcinomas. J. Natl.
Cancer Inst. 84:1486–1491.
Hope, M. J., and Cullis, P. R. 1980. Effects of divalent cations and pH on phosphatidylserine model membranes: a 31P NMR study. Biochem. Biophys. Res. Commun. 92:846–852.
Hu, F., Wang, R. Y., Hsu, T. C. 1987. Clonal origin of metastasis in B16 murine melanoma: a cytogenetic study. J. Natl. Cancer Inst. 78:155–164.
Hu, X. F., Nadalin, G., De Luise, M., Martin, T. J., Wakeling, A., Huggins, R. and Zalcberg, J.
R. 1991. Circumvention of doxorubicin resistance in multi-drug resistant human leukemia andlung cancer cells by the pure antioestrogen ICI 164384. Eur. J. Cancer 27:773–777.
Huang, S. K., Mayhew, E., Gilani, S., Lasic, D. D., Martin, F. J., and Papahadjopoulos, D. 1992.
Pharmacokinetics and therapeutics of sterically stablized liposomes in mice bearing C-26 coloncarcinoma. Cancer Res. 52:6774–6781.
Huet, S. and Robert, J. 1988. The reversal of doxorubicin resistance by verapamil is not due to an effect on calcium channels. Int. J. Cancer 41:283–286.
Inaba, M., Kobayashi, H., Sakurai, Y., and Johnson, R. R. 1979. Active efflux of daunorubicin and Adriamycin in sensitive and resistant sublines of P388 leukemia. Cancer Res. 39:2200–2203.
Inaba, M., Watanabe, T., and Sugiyama, Y. 1987. Kinetic analysis of active efflux of vincristine from multidrug-resistant P388 leukemia cells. Jpn. J. Cancer Res. 78:397–404.
Jaffr´ezou, J. P., and Laurent, G. 1993. The intriguing link between modulation of both multidrug resistance and ligand-toxin conjugate cytotoxicity. FEBS Lett. 323:191–197.
Jaffr´ezou, J. P., Levade, T., Chatelain, P., and Laurent, G. 1992. Modulation of subcellular distribu- tion of doxorubicin in multidrug-resistant P388/ADR mouse leukemia cells by the chemosen-sitizer {{2-isopropyl-1-{4-[3-N-methyl-N-(3,4-dimethoxy-b-phenethyl)amino]propyl oxy}-benzenesulfonyl}}indolizine, Cancer Res. 52:6440.
Jaffr´ezou, J. P., Herbert, J. M., Levade, T., Gau, M. N., Chatelain, P., and Laurent, G. 1991. Reversal of multidrug resistance by calcium channel blocker SR33557 without photoaffinity labeling ofP-glycoprotein, J. Biol. Chem. 266:19858.
Jain, R. K. 2008. Taming blood vessels to treat cancer. Sci. Am. 298:56–63.
2 Metastasis and Drug Resistance Jain, R. K. 1987. Transport of molecules across tumor vasculature. Cancer Metastasis Rev.
Juliano, R. L., and Ling, V. 1976. A surface glycoprotein modulating drug permeability in Chinese hamster ovary cell mutants. Biochim. Biophys. Acta 455:152–162.
Kacinski, B. M., Yee, L. D., Carter, D., Li, D., and Kuo, M. T. 1989. Human breast carcinoma cell levels of MDR-1 (P-glycoprotein) transcripts correlate in vivo inversely and reciprocally withtumor progesterone receptor content. Cancer Commun. 1:1–6.
Karsan, A., Yee, E., Poirier, G. G., Zhou, P., Craig, R., and Harlan, J. M. 1997. Fibroblast growth factor-2 inhibits endothelial cell apoptosis by Bcl-2-dependent and independent mechanisms.
Am. J. Pathol. 151:1775–1784.
Kartner, N., Riordan, J. R., and Ling, V. 1983. Cell surface P-glycoprotein associated with mul- tidrug resistance in mammalian cell lines. Science 221:1285–1288.
Kedar, D., Baker, C.H., Killion, J. J., Dinney, C. P., and Fidler, I. J. 2002. Blockade of the epidermal growth factor receptor signaling inhibits angiogenesis leading to regression of human renal cellcarcinoma growing orthotopically in nude mice. Clin. Cancer Res. 8:3592–3600.
Kellen, J. A., Wong, A., Georges, E., and Ling, V. 1991. R-verapamil decreases anti-estrogen resistance in a breast cancer model. Anticancer Res. 11:809–812.
Kerbel, R. and Folkman, J. 2002. Clinical translation of angiogenesis inhibtors. Nat. Rev. 2: Killion, J. J., Radinsky, R., and Fidler, I. J. 1999. Orthotopic models are necessary to predict therapy of transplantable tumors in mice. Cancer Metastasis Rev. 17:279–284.
Kim, S. J., Baker, C. H., Kitadai, Y., Nakamura, T., Kuwai, T., Sasaki, T., Langley, R. R., and Fidler, I.
J. The pathogenesis of cancer metastasis: relevance to therapy. In Principles of Cancer Biotherapy,5 edn., eds. R. K. Oldham, and R. O. Dillman. London: Kluwer Academic Publishers, in press.
Kim, S. J., Uehara, H., Karashima, T., Shepherd, D. L., Killion, J. J., and Fidler, I. J. 2003. Blockade of epidermal growth factor receptor signaling in tumor cells and tumor-associated endothelialcells for therapy of androgen-independent human prostate cancer growing in the bone of nudemice. Clin. Cancer Res. 9:1200–1210.
Kim, S. J., Uehara, H., Yazici, S., Busby, J. E., Nakamura, T., He, J., Maya, M., Logothetis, C., Mathew, P., Wang, X., Do, K-A., Fan, D., and Fidler, I. J. 2006. Targeting platelet-derivedgrowth factor receptor on endothelial cells of multidrug-resistant prostate cancer. J. Natl. Can-cer Inst. 98:783–793.
Kim, S. J., Uehara, H., Yazici, S., He, J., Langley, R. R., Mathew, P., Fan, D., and Fidler, I. J. 2005.
Modulation of bone microenvironment with zoledronate enhances the therapeutic effects ofSTI571 and Paclitaxel against experimental bone metastasis of human prostate Cancer. CancerRes. 65:3707–3715.
Kim, S. J., Uehara, H., Yazici, S., Langley, R. R., He, J., Tsan, R., Fan, D., Killion, J. J., and Fidler, I. J. 2004. Simultaneous blockade of platelet-derived growth factor-receptorand epidermal growth factor-receptor signaling and systemic administration of paclitaxelas therapy for human prostate cancer metastasis in bone of nude mice. Cancer Res. 64:4201–4208.
Kitadai, Y., Bucana, C. D., Ellis, L. M., Anzai, H., Tahara, E., and Fidler, I. J. 1995. In situ mRNA hybridization technique for analysis of metastasis-related genes in human colon carcinomacells. Am. J. Pathol. 147:1238–1247.
Kitadai, Y., Sasaki, T., Kuwai, T., Nakamura, T., Bucana, C. D, and Fidler, I. J. 2006. Targeting the expression of platelet-derived growth factor receptor by reactive stroma inhibits growth andmetastasis of human colon carcinoma. Am. J. Pathol. 169:2054–2065.
Klohs, W. D., Steinkampf, R. W., Havlick, M. J., and Jackson, R. C. 1986. Resistance to anthrapyra- zoles and anthracyclines in multidrug-resistant P388 murine leukemia cells: reversal by calciumblockers and calmodulin antagonists. Cancer Res. 46:4352–4356.
Kohlhardt, M., Bauer, B., Krause, H., and Fleckenstein, A. 1972. Differentiation of the transmem- brane Na and Ca channels in mammalian cardiac fibres by the use of specific inhibitors. PflugersArch. 335:309–332.
D. Fan et al.
Kroemer, H. K., Echizen, H., Heidemann, H., and Eichelbaum, M. 1992. Predictability of the in vivo metabolism of verapamil from in vitro data: Contribution of individual metabolic pathwaysand stereoselective aspects. J. Pharmacol. Exp. Ther. 260:1052–1057.
Kumar, R., Kuniyasu, H., Bucana, C. D., Wilson, M. R., and Fidler, I. J. 1998. Spatial and temporal expression of angiogenic molecules during tumor growth and progression. Oncol. Res. 10:301–311.
Kuwai, T., Nakamura, T., Kim, S. J., Sasaki, T., Kitadai, Y., Langley, R. R., Fan, D., Hamil- ton, S. R., and Fidler, I. J. 2008. Intratumoral heterogeneity for expression of tyrosine kinasegrowth factor receptors in human colon cancer surgical specimens and orthotopic tumors. Am.
J. Pathol. 172:358–366.
Lampidis, T. J., Krishan, A., Planas, L., and Tapiero, H. 1986. Reversal of intrinsic resistance to Adriamycin in normal cells by verapamil. Cancer Drug Del. 3:251–259.
Langley, R. R., Fan, D., Tsan, R. Z., Rebhun, R., He, J., Kim, S. J., and Fidler, I. J. 2004. Activation of the platelet-derived growth factor receptor enhances survival of murine bone endothelialcells. Cancer Res. (Adv. in Brief), 64:3727–3730.
Langley, R. R., Ramirez, K. M., Tsan, R. Z., Van Arsdall, M., Nilsson, M. B., and Fidler, I. J.
2003. Tissue-specific microvascular endothelial cell lines from H-2kb-tsA58 mice for studiesof angiogenesis and metastasis. Cancer Res. 63:2971–2976.
Langley, R. R., and Fidler, I. J. 2007. Tumor cell-organ microenvironment interactions in the patho- genesis of cancer metastasis. Endocr. Rev. 28:297–321.
La Porta, C. A. 2007. Drug resistance in melanoma: new perspectives. Curr. Med. Chem. 14: Lehne, G., Mørkrid, L., den Boer, M., Rugstad, H. E. 2000. Diverse effects of P-glycoprotein inhibitory agents on human leukemia cells expressing the multidrug resistance protein (MRP).
Int. J. Clin. Pharmacol. Ther. 38:187–195.
Lehnert, M., Dalton, W. S., Roe, D., Emerson, S., and Salmon, S. E. 1991. Synergistic inhibition by verapamil and quinine of P-glycoprotein-mediated multidrug resistance in a human myelomacell line model. Blood 77:348–354.
Lehnert, M., de Giuli, R., Kunke, K., Emerson, S., Dalton, W. S., and Salmon, S. E. 1996. Serum can inhibit reversal of multidrug resistance by chemosensitisers. Eur. J. Cancer 32A:862–867.
Ling, Y. H., Zou, Y. Priebe, W., and Perez-Soler, R. 1995. Partial circumvention of multi-drug resistance by annamycin is associated with comparable inhibition of DNA synthesis in thenuclear matrix of sensitive and resistant cells. Int. J. Cancer 61:402–408.
Liotta, L. A., Steeg, P. S., and Stetler-Stevenson, W. G. 1991. Cancer metastasis and angiogenesis: an imbalance of positive and negative regulation. Cell 64:327–336.
List, A. F., Spier, C., Greer, J., Azar, C., Hutter, J., Wolff, S., Salmon, S., Futscher, B., and Dalton, W., 1992. Biochemical modulation of anthracycline resistance in acute leukemia withcyclosporin-A (CSA). Proc. Am. Soc. Clin. Oncol., 11:A865.
Liu, W., Ahmad, S. A., Reinmuth, N., Shaheen, R. M., Jung, Y. D., Fan, F., and Ellis, L.
M. 2000. Endothelial cell survival and apoptosis in the tumor vasculature. Apoptosis 5:323–328.
Loor, F., Boesch, D., Gaveriaux, C., Jachez, B., Pourtier-Manzanedo, A., and Emmer, G. 1992.
SDZ 280–446, a novel semi-synthetic cyclopeptolide: in vitro and in vivo circumvention of theP-glycoprotein-mediated tumour cell multidrug resistance. Br. J. Cancer 65:11–18.
Lopez-Berestein, G., Kasi, L., Rosenblum, M. G., and Hersh, E. M. 1984. Clinical pharmacology of 99mTc-labeled liposomes in patients with cancer. Cancer Res. 44:375–378.
Lopez-Berestein, G., Fainstein, V., Hopfer, R., Mehta, K., Sullivan, M. P., Keating, M., Rosen- blum, M. G., Mehta, R., Luna, M., Hersh, E. M., Reuben, J., Juliano, R. L., and Brody, G.
P. 1985. Liposomal amphotericin B for the treatment of systemic fungal infections in patientswith cancer, a preliminary study. J. Infect. Diseases 151:704–710.
Ludwig, J. A., Szak´acs, G., Martin, S. E., Chu, B. F., Cardarelli, C., Sauna, Z. E., Caplen, N. J., Fales, H. M., Ambudkar, S.V., Weinstein, J. N., and Gottesman, M. M. 2006. Selective toxicityof NSC73306 in MDR1-positive cells as a new strategy to circumvent multidrug resistance incancer. Cancer Res. 66:4808–4815.
2 Metastasis and Drug Resistance Maeda, O., Terasawa, M., Ishikawa, T., Oguchi, H., Mizuno, K., Kawai, M., Kikkawa, F., Tomoda, Y., and Hidaka, H. 1993. A newly synthesized bifunctional inhibitor W-77, enhances Adri-amycin activity against human ovarian carcinoma cells. Cancer Res. 53:2051–2056.
Matin, K., Egorin, M. J., Ballesteros, M. F., Smith, D. C., Lembersky, B., Day, R. S., Johnson, C.
S., and Trump, D. L. 2002. Phase I and pharmacokinetic study of vinblastine and high-dosemegestrol acetate. Cancer Chemother. Pharmacol. 50:179–185.
Mazzanti, R., Croop, J. M., Gatmaitan, Z., Budding, M., Steiglitz, K., Arceci, R., and Arias, I. M.
1992. Benzquinamide inhibits P-glycoprotein mediated drug efflux and potentiates anticanceragent cytotoxicity in multidrug resistant cells. Oncol. Res. 4:359–365.
Mechetner, E. B., and Roninson, I. B. 1992. Efficient inhibition of P-glycoprotein-mediated mul- tidrug resistance with a monoclonal antibody. Proc. Natl. Acad. Sci. USA 89:5824–5828.
Mickisch, G. H., Kossig, J., Keilhauer, G., Schlick, E., Tschada, R. K., and Alken, P. M. 1990.
Effects of calcium antagonists in multidrug resistant primary human renal cell carcinomas.
Cancer Res. 50:3670–3674.
Mickisch, G. H., Merlino, G. T., Aiken, P. M., Gottesman, M. M., and Pastan, I. 1991a. New potent verapamil derivatives that reverse multidrug resistance in human renal carcinoma cells and intransgenic mice expressing the human MDR1 gene. J. Urol. 146:447–453.
Mickisch, G. H., Merlino, G. T., Galski, H., Gottesman, M. M., and Pastan, I. 1991b. Transgenic mice that express the human multidrug resistance gene in bone marrow enable a rapid identifi-cation of agents that reverse drug resistance. Proc. Natl. Acad. Sci. U.S.A. 88:547–551.
Mickisch, G. H., Pai, L. H., Gottesman, M. M., and Pastan, I. 1992. Monoclonal antibody MRK16 reverses the multidrug resistance of multidrug-resistant transgenic mice. Cancer Res. 52:4427–4432.
Miller, R. L., Bukowski, R. M., Budd, G. T., Purvis, J., Weick, J. K., Shepard, K., Midha, K. K., and Ganapathi, R. 1988. Clinical modulation of doxorubicin resistance by the calmodulin-inhibitor,trifluoperazine: a phase I/II trial. J. Clin. Oncol. 6:880–888.
Miller, T. P., Grogan, T. M., Dalton, W. S., Spier, C. M., Scheper, R. J., and Salmon, S. E. 1991.
P-glycoprotein expression in malignant lymphoma and reversal of clinical drug resistance withchemotherapy plus high-dose verapamil. J. Clin. Oncol. 9:17–24.
Millward, M. J., Cantwell, B. M. J., Lien, E. A., Carmichael, J., and Harris, A. L. 1992. Intermit- ten high-dose tamoxifen as a potential modifier of multidrug resistance. Eur. J. Cancer 28A:805–810.
Miraglia, E., Viarisio, D., Riganti, C., Costamagna, C, Ghigo, D., and Bosia, A. 2005. Na+/H+ exchanger activity is increased in doxorubicin-resistant human colon cancer cells and its mod-ulation modifies the sensitivity of the cells to doxorubicin. Int. J. Cancer 115:924–929.
Moins, N., Cayre, A., Chevillard, S., Maublant, J., Verrelle, P., and Finat-Duclos, F. 2000. Effects of MDR reversing agent combinations on the 3H-daunomycin accumulation in drug-sensitiveand drug-resistant human cancer cells. Anticancer Res. 20:2617–2623.
Morris, D. I., Speicher, L. A., Ruoho, A. E., Tew, K. D., and Seamon, K. B. 1991. Interaction of forskolin with the P-glycoprotein multidrug transporter. Biochemistry, 30:8371–8379.
Murren, J. R., Durivage, H. J., Buzaid, A. C., Reiss, M., Flynn, S. D., Carter, D., Hait, W. N.
1996. Trifluoperazine as a modulator of multidrug resistance in refractory breast cancer. CancerChemother. Pharmacol. 38:65–70.
Mutoh, K., Tsukahara, S., Mitsuhashi, J., Katayama, K., and Sugimoto, Y. 2006. Estrogen- mediated post transcriptional down-regulation of P-glycoprotein in MDR1-transduced humanbreast cancer cells. Cancer Sci. 97:1198–1204.
Nair, S. Samy, T. S., and Krishan, A. 1986. Calcium, calmodulin, and protein content of Adriamycin-resistant and -sensitive murine leukemia cells. Cancer Res. 46:229–232.
Naito, M., Yusa, K., and Tsuruo, T. 1989. Steroid hormones inhibit binding of Vinca alka- loid to multidrug resistance related P-glycoprotein. Biochem. Biophys. Res. Commun. 158:1066–1071.
Nasioulas, G., Granzow, C., Stohr, M., and Ponstingl, H. 1990. Sensitization of multidrug-resistant mouse ascites HD33 and Chinese hamster ovary CHRC5S3 cells by a photoreactive vinblastinederivative, NAPAVIN. Cancer Res. 50:403–408.
D. Fan et al.
Nawrath, H., and Raschack, M. 1987. Effects of (-)-Desmethoxy-verapamil on heart and vascular smooth muscle. J. Pharmacol. Exp. Ther. 242:1090–1097.
Nels, V., Denzer, K., and Drenchahn D. 1992. Pericyte involvement in capillary sprouting during angiogenesis in situ. Cell Tissue Res. 270:469–474.
Newman, R. A., Fan, D., Munson, H. R., Martin, L. A., and Ahmed. N. K. 1996. MDL 201,307: a novel benzothiazepine modulator of multiple drug resistance. J. Exp. Ther. Oncol. 1:109–118.
Nor, J. E., and Polverini, P. J. 1999. Role of endothelial cell survival and death signals in angio- genesis. Angiogenesis 3:101–116.
Ohnishi, S., and Ito, T. 1973. Clustering of lecithin molecules in phosphatidylserine membranes induced by calcium ion binding to phosphatidylserine. Biochem. Biophys. Res. Commun.
51:132–138.
Ozols, R. F., Cunnion, R. E., Klecker, R. W., Hamilton, T. C., Ostchega, Y., Parrillo, J. E., and Young, R. C. 1987. Verapamil and Adriamycin in the treatment of drug-resistant ovarian cancerpatients. J. Clin. Oncol. 5:641–647.
Paget, S. 1889. The distribution of secondary growths in cancer of the breast. Lancet 1:571–573.
Pajeva, I., Todorov, D. K., and Seydel, J. 2004. Membrane effects of the antitumor drugs doxoru- bicin and thaliblastine: comparison to multidrug resistance modulators verapamil and trans-flupentixol. Eur. J. Pharm. Sci. 21:243–250.
Paphadjopoulous, D. 1968. Surface properties of acidic phospholipids:interaction of monolay- ers and hydrated liquid crystals with uni- and bivalent metal ions. Biochim. Biophys. Acta163:240–254.
Pasqualini, R., Arap, W., and McDonald, D. M. 2002. Probing the structural and molecular diver- sity of tumor vasculature. Trends Mol. Med. 8:563–571.
Philip, P. A., Joel, S., Monkman, S. C., Dolega-Ossowski, E., Tonkin, K., Carmichael, J., Idle, J.
R., and Harris, A. L. 1992. A phase I study on the reversal of multidrug resistance (MDR) invivo:nifedipine plus etoposide. Br. J. Cancer 65:267–270.
Piekarz, R. L., Cohen, D., and Horwitz, S. B. 1993. Progesterone regulates the murine multidrug resistance mdr1b gene. J. Biol. Chem. 268:7613–7616.
Pirker, R., FitzGerald, D. J. P., Raschack, M., Zimmermann, F., Willingham, M. C., and Pastan, I.
1989. Enhancement of the activity of immunotoxins by analogues of verapamil. Cancer Res.
49:4791–4795.
Pirker, R., Keilhauer, G., Raschack, M., Lechner, C., and Ludwig, H. 1990. Reversal of multi- drug resistance in human KB cell lines by structural analogs of verapamil. Int. J. Cancer 45:916–919.
Poste, G., and Fidler, I. J. 1979. The pathogenesis of cancer metastasis. Nature (Lond.) 283: Racker, E. 1972. Reconstitution of a calcium pump with phospholipids and a purified Ca++- adenosine triphosphatase from sarcoplasmic reticulum, J. Biol. Chem. 247:8189.
Rahman, A., Husain, S. R., Siddiqui, J., Verma, M., Agresti, M., Center, M., Safa, A. R., and Glazer, R. I. 1992. Liposome-mediated modulation of multidrug resistance in human HL-60leukemia cells. J. Natl. Cancer Inst. 84:1909–1915.
Ramakrishnan, S., Bjorn, M. J., and Houston, L. L. 1989. Recombinant ricin A chain conjugated to monoclonal antibodies: improved tumor cell inhibition in the presence of lysosomotropiccompounds. Cancer Res. 49:613–617.
Raschko, J. W., Synold, T. W., Chow, W., Coluzzi, P., Hamasaki, V., Leong, L. A., Margolin, K. A., Morgan, R. J., Shibata, S. I., Somlo, G., Tetef, M. L., Yen, Y., ter Veer, A., andDoroshow, J. H. 2000. A phase I study of carboplatin and etoposide administered in conjunctionwith dipyridamole, prochlorperazine and cyclosporine A. Cancer Chemother Pharmacol. 46:403–410.
Reymann, A., Looft, G., Woermann, C., Dietel, M., and Erttmann, R. 1993. Reversal of mul- tidrug resistance in Friend leukemia cells by dexniguldipine HCl. Cancer Chemother. Pharma-col. 32:25–30.
2 Metastasis and Drug Resistance Risau, W., Drexler, H., Mironov, V., Smits, A., Siegbahn, A., Funa, K. and Heldin, C. H. 1992.
Platelet-derived growth factor is angiogenic in vivo. Growth Factors 7:261–266.
Rougier, O., Vossort, G., Garnier, D., Gargouil, Y. M., and Coraboeuf, E. 1969. Existence and role of a slow inward current during the frog atrial action potential. Pflugers Arch. 308:91–110.
Ruiz, J. C., Choi, K. H., Von Hoff, D. D., Robinson, I. B., and Wahl, G. M. 1989. Autonomously replicating episomes contain mdr1 genes in a multidrug-resistant human cell line. Mol. CellBiol. 9:109–115.
Safa, A. R., Glover, C. J., Sewell, J. L., Meyers, M. B., Biedler, J. L., and Felsted, R. L. 1987.
Identification of the multidrug resistance-related membrane glycoprotein as an acceptor forcalcium channel blockers. J. Biol. Chem. 262:7884–7888.
Safa, A. R., Agresti, M., Tamai, I., Mehta, N. D. and Vahabi, S. 1990. The alpha 1-adrenergic photoaffinity probe 125I:arylazidoprazosin binds to a specific peptide of P-glycoprotein inmultidrug-resistant cells. Biochem. Biophys. Res. Commun. 166:259–266.
Salmon, S. E., Dalton, W. S., Grogan, T. M., Plezia, P., Lehnert, M., Roe, D. J., and Miller, T. P.
1991. Multidrug resistant myeloma: laboratory and clinical effects of verapamil as a chemosen-sitizer. Blood 78:44–50.
Saltz, L., Murphy, B., Kemeny, N., Bertino, J., Tong, W., Keefe, D., Tzy-Jun, Y., Tao, Y., Kelsen, D., OBrien, J. P. 1994. A phase I trial of intrahepatic verapamil and doxorubicin. Regionaltherapy to overcome multidrug resistance. Cancer 74:2757–2764.
Saponara, S., Ferrara, A, Gorelli, B., Shah, A., Kawase, M., Motohashi, N., Molnar, J., Sgaragli, G., and Fusi, F. 2007. 3,5-dibenzoyl-4-(3-phenoxyphenyl)-1,4-dihydro-2,6-dimethylpyridine(DP7): a new multidrug resistance inhibitor devoid of effects on Langendorff-perfused rat heart.
Eur. J. Pharmacol. 563:160–163.
Sarkadi, B., Price, E. M., Boucher, R. C., Germann, U. A., and Scarborough, G. A. 1992. Expres- sion of the human multidrug resistance cDNA in insect cells generates a high activity drug-stimulated membrane ATPase. J. Biol. Chem. 267:4854–4858.
Sasaki, T., Kitadai, Y., Nakamura, T., Kim, J. S., Tsan, R. Z., Kuwai, T., Langley, R. R., Fan, D., Kim, S. J., and Fidler, I. J. 2007. Inhibition of epidermal growth factor receptor and vascularendothelial growth factor receptor phosphorylation on tumor-associated endothelial cells leadsto treatment of orthotopic human colon cancer in nude mice. Neoplasia 9:1066–1077.
Sato, W., Fukazawa, N., Suzuki, T., Yusa, K., and Tsuruo, T. 1991. Circumvention of multidrug resistance by a newly synthesized quinoline derivative, MS-073. Cancer Res. 51:2420–2424.
Schackert, G., Fan, D., Nayar, R., and Fidler, I. J. 1989. Arrest and retention of multilamellar liposomes in the brain of normal mice or mice bearing experimental brain metastases. Sel.
Cancer Ther. 5:73–79.
Schr¨oder, J., Esteban, M., M¨uller, M. R., Kasimir-Bauer, S., Bamberger, U., Heckel, A., Seeber, S., and Scheulen, M. E. 1996. Modulation of multidrug resistance by BIBW22BS in blasts ofde novo or relapsed or persistent acute myeloid leukemia ex vivo. J. Cancer Res. Clin. Oncol.
122:307–312.
Schuurhuis, G. J., Broxterman, H. J., van der Hoeven, J. J. M., Pinedo, H. M., and Lankelma, J. 1987. Potentiation of doxorubicin cytotoxicity by the calcium antagonistbepridil in anthracycline-resistant and -sensitive cell lines. Cancer Chemother. Pharmacol. 20:285–290.
Seid, C. A., Fidler, I. J., Clyne, R. K., Earnest, L. E., and Fan, D. 1991. Overcoming murine tumor cell resistance to vinblastine by presentation of the drug in multilamellar liposomes consistingof phosphatidylcholine and phosphatidylserine. Sel. Cancer Ther. 7:103–112.
Sessa, C., Calabresi, F., Cavalli, F., Cerny, T., Liati, P., Skovsgaard, T., Sorio, R., and Kaye, S. B.
1992. Phase II studies of 4-iodo-4-deoxydoxorubicin in advanced non-small cell lung, colonand breast cancers. Ann. Oncol. 2:727–731.
Shen, F., Chu, S., Bence, A. K., Bailey, B., Xue, X., Erickson, P. A., Montrose, M. H., Beck, W. T., and Erickson, L. C. 2008. Quantitation of doxorubicin uptake, efflux, and modulationof multidrug resistance (MDR) in MDR human cancer cells. J. Pharmacol. Exp. Ther. 324:95–102.
D. Fan et al.
Singh, R. K., Bucana, C. D., Gutman, M., Fan D., Wilson, M., and Fidler, I. J. 1994. Organ site- dependent expression of basic fibroblast growth factor in human renal cell carcinoma cells. Am.
J. Pathol. 145:365–374.
Singh, R. K., Gutman, M., Bucana, C. D., Sanchez, R., Llansa, N., and Fidler, I. J. 1995. Inter- ferons alpha and beta downregulate the expression of basic fibroblast growth factor in humancarcinomas. Proc. Natl. Acad. Sci. USA 92:4562–4566.
Singh, R. K., Gutman, M., Radinsky, R., Bucana, C. D., and Fidler, I. J. 1994. Expression of interleukin-8 correlates with the metastatic potential of human melanoma cells in nude mice.
Cancer Res. 54:3242–3247.
Sinicrope, F. A., Dudeja, P. K., Bissonnette, B. M., Safa, A. R., and Brasitus, T. A., 1992. Mod- ulation of P-glycoprotein-mediated drug transport by alterations in lipid fluidity of rat livercanalicular membrane vesicles. J. Biol. Chem. 267:24995–25002.
Slater, L. M., Murray, S. L., Wetzel, M. W., Wisdom, R. M., and DuVall, E. M. 1982. Verapamil restoration of daunorubicin responsiveness in daunorubicin-resistant Ehrlich ascites carcinoma.
J. Clin. Invest. 70:1131–1134.
Slater, L. M., Sweet, P., Stupecky, M., and Gupta, S. 1986a. Cyclosporin A reverses vin- cristine daunorubicin resistance in acute lymphatic leukemia in vitro. J. Clin. Invest. 77:1405–1408.
Slater, L. M., Sweet, P., Stupecky, M., Wetzel, M. W. and Gupta, S. 1986b. Cyclosporin A corrects daunorubicin resistance in Ehrlich ascites carcinoma. Br. J. Cancer 54:235–238.
Solary, E., Caillot, D., Chauffert, B., Casasnovas, R. O., Dumas, M., Maynadie, M., and Guy, H.
1992. Feasibility of using quinine, a potential multidrug resistance-reversing agent, in combi-nation with mitoxantrone and cytarabine for the treatment of acute leukemia. J. Clin. Oncol.,10:1730–1736.
Sotelo, J., Brice˜no, E., and L´opez-Gonz´alez, M. A. 2006. Adding chloroquine to conventional treatment for glioblastoma multiforme: a randomized, double-blind, placebo-controlled trial.
Ann. Intern. Med. 144:337–43.
Spoelstra, E. C., Dekker, H., Schuurhuis, G. J., Broxterman, H. J., and Lankelma, J. 1991. P- glycoprotein drug efflux pump involved in the mechanisms of intrinsic drug resistance in vari-ous colon cancer cell lines. Evidence for a saturation of active daunorubicin transport. Biochem.
Pharmacol. 41:349–359.
Sridhar, K. S., Krishan, A., Samy, T. S. A., Sauerteig, A., Wellham, L. L., McPhee, G., Duncan, R. C., Anac, S. Y., Ardalan, B., and Benedetto, P. W. 1993. Prochlorperazine as a doxorubicin-efflux blocker:phase I clinical and pharmacokinetics studies. Cancer Chemother. Pharmacol.
31:423–430.
Stein, Y., and Stein, O. 1966. Metabolism of labeled lysolecithin, lysophosphatidylethanolamine and lecithin in the rat. Biochim. Biophys. Acta 116:95–107.
Stuart, N. S., Philip, P., Harris, A. L., Tonkin, K., Houlbrook, S., Kirk, J., Lien, E. A., and Carmichael, J. 1992. High-dose tamoxifen as an enhancer of etoposide cytotoxicity. Clini-cal effects and in vitro assessment in P-glycoprotein expressing cell lines. Br. J. Cancer 66:833–839.
Suhardja, A., and Hoffman, H. 2003. Role of growth factors and their receptors in proliferation of Microvascular endothelial cells. Microsc. Res. Tech. 60:70–75.
Takahashi, Y., Mai, M., Wilson, M. R., and Ellis, L. M. 1996. Site-dependent expression of vascular endothelial growth factor, angiogenesis, and proliferation in human gastric carcinoma. Int. J.
Oncol. 8:701–705.
Talmadge, J. E., Wolman, S. R., and Fidler, I. J. 1982. Evidence for the clonal origin of spontaneous metastases. Science 217:361–363.
Teodori, E., Dei, S., Garnier-Suillerot, A., Gualtieri, F., Manetti, D., Martelli, C., Romanelli, M. N, Scapecchi, S., Sudwan, P., and Salerno, M. 2005. Exploratory chemistry toward theidentification of a new class of multidrug resistance reverters inspired by pervilleine and vera-pamil models. J. Med. Chem. 48:7426–36.
2 Metastasis and Drug Resistance Thierry, A. R., Rahman, A., and Dritschilo, A. 1993. Overcoming multidrug resistance in human tumor cells using free and liposomally encapsulated antisense oligodeoxynucleotides.
Biochem. Biophys. Res. Commun. 190:952–960.
Thierry, A. R., Vige, D., Coughlin, S. S., Belli, J. A., Dritschilo, A., and Rahman, A. 1993. Modula- tion of doxorubicin resistance in multidrug-resistant cells by liposomes. FASEB J. 7:572–579.
Todorov, D. K. 1988. Thaliblastine. Drug Future 13:234–238.
Tr¨auble, H., and Eibl, H. 1974. Electrostatic effects on lipid phase transitions:membrane structure and ionic environment. Proc. Natl. Acad. Sci. USA 71:214–219.
Trump, D. L., Smith, D. C., Ellis, P. G., Rogers, M. P., Schold, S. C., Winer, E. P., Panella, T. J., Jordan, V. C., and Fine, R. L., 1992. High-dose oral tamoxifen, a potential multidrug-resistance-reversal agent:phase I trial in combination with vinblastine. J. Natl. Cancer Inst. 84:1811–1816.
Tsuruo, T., Iida, H. Kitani, Y., Yokota, K., Tsukaggoshi, S., and Sukurai, Y. 1984. Effects of quini- dine and related compounds on cytoxicity and cellular accumulation of vincristine and Adri-amycin in drug-resistant tumor cells. Cancer Res. 44:4303–4307.
Tsuruo, T., Iida, H., Nojiri, M., Tsukagoshi, S., and Sakurai, Y. 1983a. Potentiation of vincristine and Adriamycin in human hematopoietic tumor cell lines by calcium antagonists and calmod-ulin inhibitors. Cancer Res. 43:2267–2272.
Tsuruo, T., Iida, H., Nojiri, M., Tsukagoshi, S., and Sakurai, Y. 1983b. Circumvention of vin- cristine and Adriamycin resistance in vitro and in vivo by calcium influx blockers. Cancer Res.
43:2905–2910.
Tsuruo, T., Iida, H., Tsukagoshi, S., and Sakurai, Y. 1981. Overcoming of vincristine resistance in P388 leukemia in vivo and in vitro through enhanced cytotoxicity of vincristine and vinblastineby verapamil. Cancer Res. 41:1967–1972.
Tsuruo, T., Iida, H., Tsukagoshi, S., and Sakurai, Y. 1982. Increased accumulation of vincristine and Adriamycin in drug-resistant P388 tumor cells following incubation with calcium antago-nists and calmodulin inhibitors. Cancer Res. 42:4730–4733.
Twentyman, P. R., Fox, N. E., and White, D. J. G. 1987. Cyclosporine A and its analogues as modifiers of Adriamycin and vincristine resistance in a multidrug resistant human cancer cellline. Br. J. Cancer 56:55–57.
Uehara, H., Kim, S. J., Karashima, T., Shepherd, D. L., Fan, D., Tsan, R., Killion, J. J., Logo- thetis, C., Mathew, P., and Fidler, I. J. 2003. Effects of blocking platelet-derived growth factor-receptor signaling in a mouse model of prostate cancer bone metastasis. J. Natl. Cancer Inst. 95:458–470.
Venditti, J. M. 1981. Preclinical drug development: rationale and methods. Semin. Oncol. 8: Verstuyft, C., Strabach, S., El-Morabet, H., Kerb, R., Brinkmann, U., Dubert, L., Jaillon, P., Funck- Brentano, C., Trugnan, G., and Becquemont, L. 2003. Dipyridamole enhances digoxin bioavail-ability via P-glycoprotein inhibition. Clin. Pharmacol. Ther. 73:51–60.
Verweij, J., Herweijer, H., Oosterom, R., van der Burg, M. E., Planting, A. S., Seynaeve, C., Stoter, G., and Nooter, K. 1992. A phase II study of epidoxorubicin in colorectal cancer and the use ofcyclosporin-A in an attempt to reverse multidrug resistance. Br. J. Cancer 64:361–364.
Vezmar, M., and Georges, E. 2000. Reversal of MRP-mediated doxorubicin resistance with quinoline-based drugs. Biochem. Pharmacol. 59:1245–1252.
Vickers, P. J., Dickson, R. B., Shoemaker, R., and Cowan, K. H. 1988. A multidrug-resistant MCF- 7 human breast cancer cell line which exhibits cross-resistance to anti-estrogens and hormone-independent tumor growth in vivo. Mol. Endocrinol. 2:886–892.
Von Hoff, D. D., Forseth, B., Clare, C. N., Hansen, K. L., and VanDevanter, D. 1990. Double minutes arise from circular extrachromosomal DNA intermediates which integrate into chro-mosomal sites in human HL-60 leukemia cells. J. Clin. Invest. 85:1887–1895.
Wadler, S., and Wiernik, P. H. 1988. Partial reversal of doxorubicin resistance by forskolin and 1,9-dideoxyforskolin in murine sarcoma S180 variants. Cancer Res. 48:539–543.
D. Fan et al.
Wang, J. H., Wu, Q. D., Bouchier-Hayes, D., and Redmond, H. P. 2002. Hypoxia upregulates Bcl-2 expression and suppresses interferon-gamma-induced antiangiogenic activity in humantumor-derived endothelial cells. Cancer 15:2745–2755.
Weidner, N., Folkman, J., Pozza, F., Bevilacqua, P., Allred, E. N., Moore, D. H., Meli, S., and Gasparini, G. 1992. Tumor angiogenesis:a new significant and independent prognostic indicatorin early-stage breast carcinoma. J. Natl. Cancer Inst. 84:1875–1887.
Wilmanns, C., Fan, D., OBrian, C. A., Bucana, C. D., and Fidler, I. J. 1992. Orthotopic and ectopic organ environments differentially influence the sensitivity of murine colon carcinoma cells todoxorubicin and 5-fluorouracil. Int. J. Cancer 52:98–104.
Wilson, C. M., Serrano, A. E., Wasley, A., Bogenschutz, M. P., Shankar, A. H. and Wirth, D. F.
1989. Amplification of a gene related to mammalian mdr genes in drug-resistant Plsmodiumfaciparum. Science 244:1184–1186.
Woodcock, D. M., Jefferson, S., Linsenmeyer, M. E., Crowther, P. J., Chojnowski, G. M., Williams, B. and Bertoncello, I. 1990. Reversal of the multidrug resistance phenotype with CremophorEL, a common vehicle for water-insoluble vitamins and drugs. Cancer Res. 50:4199–4203.
Woodcock, D. M., Linsenmeyer, M. E., Chojnowski, G., Kriegler, A. B., Nink, V., Webster, L.
K., and Sawyer, W. H. 1992. Reversal of multidrug resistance by surfactants. Br. J. Cancer66:62–68.
Wu, H., Hait, W. N., and Yang, J. M. 2003. Small interfering RNA-induced suppression of MDR1 (P-glycoprotein) restores sensitivity to multidrug-resistant cancer cells. Cancer Res. 63:1515–1519.
Wurz, G. T., Emshoff, V. D., Degregorio, M. W., and Wiebe, V. J. 1993. Targeting chemosensitizing doses of toremifene based on protein binding. Cancer Chemother. Pharmacol. 31:412–414.
Yang, C. P., Cohen, D., Greenberger, L. M., Hsu, S. I., and Horwitz, S. B. 1990. Differential transport properties of two mdr gene products are distinguished by progesterone. J. Biol. Chem.
265:10282–10288.
Yang, C. P., DePinho, S. G., Greenberger, l. M., Arceci, R. J., and Horwitz, S. B. 1989. Proges- terone interacts with P-glycoprotein in multidrug-resistant cells and in the endometrium ofgravid uterus. J. Biol. Chem. 264:782–788.
Yin, Y., Allen, P. D., Jia, L., MacEy, M. G., Kelsey, S. M., and Newland, A. C. 2000. Constitutive levels of cAMP-dependent protein kinase activity determine sensitivity of human multidrug-resistant leukaemic cell lines to growth inhibition and apoptosis by forskolin and tumour necro-sis factor alpha. Br. J. Haematol. 108:565–573.
Yokoi, K,, Sasaki, T., Bucana, C. D., Fan, D., Baker, C. H., Kitadai, Y., Kuwai, T., Abbruzzese, J. L., and Fidler, I. J. 2005. Simultaneous inhibition of EGFR, VEGFR, and platelet-derivedgrowth factor receptor signaling combined with gemcitabine produces therapy of human pan-creatic carcinoma and prolongs survival in an orthotopic nude mouse model. Cancer Res.
65:10371–10380.
Yoneda, J., Kuniyasu, H., Crispens, M. A., Price, J. E., Bucana, C. D., and Fidler, I. J. 1998. Expres- sion of angiogenesis-related genes and progression of human ovarian carcinomas in nude mice.
J. Natl. Cancer Inst. 90:447–454.
Yu, J.L., Rak, J. W., Carmeliet, P., Nagy, A., Kerbel, R. S., and Coomber, B. L. 2001. Heteroge- neous vascular dependence of tumor cell populations. Am. J. Pathol. 158:1225–1234.
Zalipsky, S., Saad, M., Kiwan, R., Ber, E., Yu, N., and Minko, T. 2007. Antitumor activity of new liposomal prodrug of mitomycin C in multidrug resistant solid tumor:insights of the mechanismof action. J. Drug Target 15:518–530.
Zhu, H. J., Wang, J. S., Guo, Q. L., Jiang, Y., and Liu, G. Q. 2005. Reversal of P-glycoprotein mediated multidrug resistance in K562 cell line by a novel synthetic calmodulin inhibitor, E6.
Biol. Pharm. Bull. 10:1974–1978.

Source: http://beck-shop.de/fachbuch/leseprobe/9780387894447_excerpt_001.pdf

Untitled

Antibiotic Therapy vs Appendectomy for Treatmentof Uncomplicated Acute AppendicitisThe APPAC Randomized Clinical Trial Paulina Salminen, MD, PhD; Hannu Paajanen, MD, PhD; Tero Rautio, MD, PhD; Pia Nordström, MD, PhD; Markku Aarnio, MD, PhD;Tuomo Rantanen, MD, PhD; Risto Tuominen, MPH, PhD; Saija Hurme, MSc; Johanna Virtanen, MD; Jukka-Pekka Mecklin, MD, PhD;Juhani Sand, MD, PhD; Airi Jartti, MD; Irina Rinta-Kiikka, MD, PhD; Juha M. Grönroos, MD, PhD

Pone.0113936 1.15

Randomized Placebo-Controlled Phase IITrial of Autologous Mesenchymal StemCells in Multiple Sclerosis Sara Llufriu1., Marı´a Sepu´lveda1., Yolanda Blanco1, Pedro Marı´n2,Beatriz Moreno1, Joan Berenguer3, In˜igo Gabilondo1, Eloy Martı´nez-Heras1,Nuria Sola-Valls1, Joan-Albert Arnaiz4, Enrique J. Andreu5, Begon˜a Ferna´ndez1,Santi Bullich1, Bernardo Sa´nchez-Dalmau1,6, Francesc Graus1, Pablo Villoslada1,Albert Saiz1*

Copyright © 2008-2016 No Medical Care