selective delivery of therapeutic agents for the diagnosis and treatment of cancer

16
Review 10.1517/14712598.6.1.39 © 2006 Ashley Publications ISSN 1471-2598 39 Ashley Publications www.ashley-pub.com Delivery Selective delivery of therapeutic agents for the diagnosis and treatment of cancer Girja S Shukla & David N Krag Vermont Comprehensive Cancer Center, Department of Surgery, University of Vermont College of Medicine, 89 Beaumont Avenue, Burlington, VT 05405, USA Research activity aimed towards achieving specific and targeted delivery of cancer therapeutics has expanded tremendously in the last decade, resulting in new ways of directing drugs to tumours, as well as new types of drugs. The available strategies exploit differences in the nature of normal and cancer cells and their microenvironment. The discovery and validation of cancer-associated markers, as well as corresponding ligands, is pivotal for developing selective delivery technology for cancer. Although most current clinical trials are either monoclonal antibody- or gene-based, methodological advances in combinatorial libraries of peptides, single chain variable fragments and small organic molecules are expected to change this scenario in the near future. Nanotechnology platforms today allow systematic and modular combinations of therapeutic agents and tumour-binding moieties that may generate novel, personalised agents for selective delivery in cancer. This paper discusses recent developments and future prospects of targeted delivery technologies in the management of cancer. Keywords: ADEPT, cancer-specific targets, GDEPT, ligands, molecular diagnosis, prodrug therapy, targeted delivery, tumour imaging Expert Opin. Biol. Ther. (2006) 6(1):39-54 1. Introduction Delivery of anticancer drugs through veins is essentially uncontrolled delivery of drugs to all cells of the body, exposing normal cells as well as cancer cells to the nonspecific antiproliferative effects of anticancer drugs. The majority of existing cancer chemotherapeutics are delivered in this manner. What little selectivity the chemotherapeutics have for the cancer cells is due to the greater sensitivity of cancer cells. The success of these cancer drugs is limited by concentration-dependent systemic toxicity. The inability to deliver an adequate antiproliferative concentration of drug results in failure to eliminate all of the cancer cells and the situation is worsened by the subsequent development of drug resistance [1]. Developing strategies that help to restrict the action of therapeutic agents to cancer cells are expected to decrease systemic toxicity and increase the efficacy of cancer cell killing. The development of procedures for selective delivery of therapeutic agents for the diagnosis and treatment of cancer is an active area of ongoing investigations in both experimental and clinical trials. In broad terms, selective delivery of cancer therapeutic agents includes any methodology by which the functional concentration of drug is higher at the cancer site than in normal tissue. A wide variety of methods may fall under the category of ‘selective delivery’, including interventions as simple and mechanical as selective vascular administration in which the drug is physically isolated in a tumour-bearing area. An example is isolated limb perfusion, in which the injected drug is confined to a localised region of the body [2]. The drug 1. Introduction 2. Selective delivery by passive targeting 3. Selective delivery through cancer-specific antibodies 4. Peptide-mediated selective delivery 5. Selective delivery through nucleotide aptamers 6. Selective delivery by prodrug activation 7. External energy-controlled delivery 8. Organ-selective delivery 9. Expert opinion and conclusion For reprint orders, please contact: [email protected] Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by Universiteit Twente on 11/20/14 For personal use only.

Upload: david-n

Post on 27-Mar-2017

212 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Selective delivery of therapeutic agents for the diagnosis and treatment of cancer

Review

10.1517/14712598.6.1.39 © 2006 Ashley Publications ISSN 1471-2598 39

Ashley Publicationswww.ashley-pub.com

Delivery

Selective delivery of therapeutic agents for the diagnosis and treatment of cancerGirja S Shukla† & David N Krag†Vermont Comprehensive Cancer Center, Department of Surgery, University of Vermont College of Medicine, 89 Beaumont Avenue, Burlington, VT 05405, USA

Research activity aimed towards achieving specific and targeted delivery ofcancer therapeutics has expanded tremendously in the last decade, resultingin new ways of directing drugs to tumours, as well as new types of drugs. Theavailable strategies exploit differences in the nature of normal and cancercells and their microenvironment. The discovery and validation ofcancer-associated markers, as well as corresponding ligands, is pivotal fordeveloping selective delivery technology for cancer. Although most currentclinical trials are either monoclonal antibody- or gene-based, methodologicaladvances in combinatorial libraries of peptides, single chain variablefragments and small organic molecules are expected to change this scenarioin the near future. Nanotechnology platforms today allow systematic andmodular combinations of therapeutic agents and tumour-binding moietiesthat may generate novel, personalised agents for selective delivery in cancer.This paper discusses recent developments and future prospects of targeteddelivery technologies in the management of cancer.

Keywords: ADEPT, cancer-specific targets, GDEPT, ligands, molecular diagnosis, prodrug therapy, targeted delivery, tumour imaging

Expert Opin. Biol. Ther. (2006) 6(1):39-54

1. Introduction

Delivery of anticancer drugs through veins is essentially uncontrolled delivery ofdrugs to all cells of the body, exposing normal cells as well as cancer cells to thenonspecific antiproliferative effects of anticancer drugs. The majority of existingcancer chemotherapeutics are delivered in this manner. What little selectivity thechemotherapeutics have for the cancer cells is due to the greater sensitivity of cancercells. The success of these cancer drugs is limited by concentration-dependentsystemic toxicity. The inability to deliver an adequate antiproliferative concentrationof drug results in failure to eliminate all of the cancer cells and the situation isworsened by the subsequent development of drug resistance [1]. Developingstrategies that help to restrict the action of therapeutic agents to cancer cells areexpected to decrease systemic toxicity and increase the efficacy of cancer cell killing.

The development of procedures for selective delivery of therapeutic agents for thediagnosis and treatment of cancer is an active area of ongoing investigations in bothexperimental and clinical trials. In broad terms, selective delivery of cancertherapeutic agents includes any methodology by which the functional concentrationof drug is higher at the cancer site than in normal tissue. A wide variety of methodsmay fall under the category of ‘selective delivery’, including interventions as simpleand mechanical as selective vascular administration in which the drug is physicallyisolated in a tumour-bearing area. An example is isolated limb perfusion, in whichthe injected drug is confined to a localised region of the body [2]. The drug

1. Introduction

2. Selective delivery by

passive targeting

3. Selective delivery through

cancer-specific antibodies

4. Peptide-mediated

selective delivery

5. Selective delivery through

nucleotide aptamers

6. Selective delivery by

prodrug activation

7. External energy-controlled

delivery

8. Organ-selective delivery

9. Expert opinion and conclusion

For reprint orders, please contact:[email protected]

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

iteit

Tw

ente

on

11/2

0/14

For

pers

onal

use

onl

y.

Page 2: Selective delivery of therapeutic agents for the diagnosis and treatment of cancer

Selective delivery of therapeutic agents for the diagnosis and treatment of cancer

40 Expert Opin. Biol. Ther. (2006) 6(1)

concentration can be considerably increased with correspond-ingly increased effectiveness of killing cancer cells. While thisapproach supports the notion that an increased concentrationof drug can lead to increased killing of cancer cells, any canceroutside the limb remains untreated. Most strategies, however,are pharmaceutical and are the subject of this review. In theseapproaches, the differences in the biochemical and physio-logical nature of normal and cancer cells and their micro-environment are exploited for selective delivery [3,4]. Not allpharmaceutical strategies of selective delivery fit into neatcategories, but a common feature is to increase the effectiveconcentration of bioactive molecules at the target site. Thesestrategies include:

• use of drug carriers that, due to their physical properties,accumulate preferentially at the tumour

• use of ligands that bind to overexpressed tumour-associatedantigens

• activation of anticancer drugs preferentially at the cancer site• external energy irradiation to release drugs at the cancer site

In recent years, the overexpressing cancer-specific antigens havebecome an important tool in developing different deliverytechnologies in cancer. The delivery by targeting cancer-specificantigens concentrates the therapeutics at the cancer site, andthis helps in diagnostic imaging and treatment of cancers. Forexample, ∼ 2,000,000 HER-2 receptors are present on anoverexpressing cancer cell surface, whereas a non-overexpress-ing normal cell has 10,000 – 20,000 molecules [5]. Thisrepresents ∼ 100-fold higher drug concentrations reaching tocancer cells than normal cells, when delivered through aHER-2-binding ligand.

A variety of technologies using combinations of differentapproaches are constantly being developed for selective deliv-ery of therapeutics in cancer diagnosis and treatment [6,7].These delivery systems employ different targets (e.g., cancercell and neovascular antigens, hypoxia, high osmoticpressure), targeting agents (e.g., monoclonal antibodies[mAbs], single chain variable fragments [scFvs], peptides,oligonucleotides), effectors (e.g., chemical or biologicaltoxins, radioisotopes, genes, enzymes, immunomodulators,oligonucleotides, imaging and diagnostic agents), vehicles(e.g., colloidal systems, including liposomes, emulsions,micelles, nanoparticles; polymer conjugates or implants;viruses; bacteria; modified allogenic cells), and drug-releasingswitches (e.g., thermal, radiation, ultrasound, magnetic field,visible or UV light, enzymes). This paper presents recentdevelopments in the field, with particular reference to deliverysystems that use cancer-targeting mechanisms for deliveringtherapeutics in cancer diagnosis and treatment.

2. Selective delivery by passive targeting

A therapeutic agent stably associated with sustained-release drugcarriers, such as liposomes, polymers and other nanoparticles,has been used for selective delivery of therapeutics for diagnosis

and treatment in cancer. These carriers have been shown toextravasate and localise to areas of increased vascular permeabil-ity, such as those found in angiogenic tumours, in a processreferred to as passive targeting. Of these drug delivery systems,liposome-based formulations of the anthracycline drugs havehad a great impact on the treatment of cancer patients to date.Tumour vessels have an increased permeability to macromole-cules owing to a lack of tight junctions that are normally presentbetween adjacent vasculature endothelial cells of other vessels.The size of the gaps between the cells in tumour vessels is suffi-ciently large to allow the extravasation of most targetedtherapeutics from vessels into the tumour interstitial space [6-8].The therapeutic agents retained in these carriers while in circula-tion are not freely available, thus minimising the uptake bynormal tissues. Surface modification of carriers with polyethyl-ene glycol (PEG), commonly referred to as the pegylationprocess, to produce sterically hindered or STEALTH liposomes,has been shown to protect carriers from reticuloendothelialsystem uptake [7]. This allows extended circulation time, whichin turn maximises carrier uptake by tumour. The limitedlymphatic drainage in tumours helps the carrier-bound drug toaccumulate in tumour tissue. The phenomenon of thecombined effect of these factors is termed the enhanced permea-tion and retention (EPR) effect. The pegylated liposomal doxo-rubicin is in the market and currently being used for cancertreatment. Recently, composite vehicle particles of 80 − 120 nm,known as ‘nanocells’, consisting of a solid biodegradable polymercore surrounded by a pegylated-lipid membrane, were used forstaged delivery of two drugs in the treatment of cancer [9]. Theantiangiogenic drug combretastatin dissolved in a lipid envelopewas released first, followed by a slow release of the chemothera-peutic drug doxorubicin from polymer degradation. The firstdelivery of the antiangiogenic factor led to tumour vasculatureshut down, trapping in the tumour the inner particle, withsecond payload of chemotherapeutic drug. The subsequentrelease of the chemotherapy in the tumour killed the cancer cells.This is an example of next-generation colloidal delivery systemsaiming for an integrative approach in cancer treatment. Table 1shows some of the colloidal system-based anticancer drugsavailable in the market for clinical management of cancer.

Passive targeting by extravasation from tumour vesselsallows drug-encapsulating carriers to accumulate withintumour stroma and release the bioactive drug after enzymaticdegradation and/or phagocytic attack, with subsequent diffu-sion into the cancer cells. Recent developments in this fieldhave generated systems that deliver the therapeutic directly tocancer cells through active tumour targeting. This has beenachieved by linking therapeutic moieties to high-affinityligands directed against cancer-associated targets.

3. Selective delivery through cancer-specific antibodies

The advent of mAb technology in the 1970s and the develop-ment of genetically engineered derivatives in the 1980s, along

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

iteit

Tw

ente

on

11/2

0/14

For

pers

onal

use

onl

y.

Page 3: Selective delivery of therapeutic agents for the diagnosis and treatment of cancer

Shukla & Krag

Expert Opin. Biol. Ther. (2006) 6(1) 41

with technological advances in the bulk production of mAbs,have led to a number of clinical studies to evaluate the efficacyof cancer-specific mAbs in the targeted delivery in cancer.Several mAbs have been approved in recent years and are inthe market for their use in cancer management (Table 2). Theexquisite specificity of mAbs makes them ideal vehicles forselective delivery of a variety of diagnostic/imaging agents orbiologically active molecules, such as chemotherapy, radioiso-topes, immunomodulators, chemical and biological toxins,and fluorescence molecules [10-17]. Gemtuzumab ozogamicinis the first mAb that was approved for selective delivery of asmall molecule cytotoxin to cancer cells. The antibody islinked with the highly potent DNA-cleaving agent calicheam-icin through an acid-labile hydrazone moiety, which, follow-ing internalisation into low pH endosomes, breaks andreleases a free cytotoxic agent. Several recombinant immuno-toxin products carrying Pseudomonas exotoxin, such asLMB-2 (anti-CD25) and BL22 (anti-CD22), are in cancerclinical trials. Ibritumomab tiuxetan and tositumomabantibodies that deliver radiotoxic agents to cancer cells areapproved for the treatment of non-Hodgkin’s lymphoma.Another mAb delivering radioisotopes, labetuzumab(CEA-Cide™, anticarcinoembronic antigen), is in clinicaltrials in colorectal and pancreatic cancer patients. Antibodieshave also been employed for delivering magnetic resonanceimaging (MRI) contrast agents [18-20], radionuclides [21,22],quantum dots [8] and metal-based nanoshells [23] for cancerimaging. The naked or unlabelled antibodies that are already inthe market for cancer treatment (Table 2) and those being evalu-ated in cancer clinical trials at present(e.g., 17-1A [edrecolomab,anti-EpCAM]; B43.13 [oregovomab, anti-CA125]; epratuzu-mab [anti-CD22]; and Hu1D10 [apolizumab, anti-1D10])have potential for use as a vehicles for selective delivery oftherapeutic agents in cancer diagnosis and treatment.

Although encouraging results with antibody conjugates inthe management of haematopoietic malignancies, especiallynon-Hodgkin’s lymphoma, have generated considerableinterest in such therapy, experience with solid tumours hasbeen less enthusiastic [10,24]. This is due, at least in part, to avariety of factors, such as poor tumour vascularisation, barriersto antibody penetration and high intratumoural pressure, thatlimit the antibody conjugate delivery in solid tumours incomparison to the relative accessibility of the malignant cells inblood and bone marrow. The major limitations in antibody-targeted delivery are related to large antibody size, nonspecificuptake of antibodies by the liver and the reticuloendothelialsystem, and immunogenicity [24]. The development of humanor chimeric antibodies has largely overcome the host immuneresponses to rodent-derived mAbs [25]. The use of antibodyfragments, such as Fab, F(ab’)2 and scFv, has been helpful toresolve certain size-related issues [24,26,27]. In a clinical trial, thescFv MFE-23 against carcinoembryonic antigen has been usedfor selective delivery of a radioactive tracer for gamma-cameraimaging of tumour [28]. The development of phage displaytechnology for the selection of target-specific scFvs has openedenormous possibilities in this field [29,30].

4. Peptide-mediated selective delivery

Carrier peptides, acting as shuttles for the selective delivery ofanticancer compounds, are much smaller than antibodies, butstill retain selective binding affinity to cancer target molecules,and may overcome several of the limitations of antibodytherapy [31-33]. One of the great strengths of peptidesas potential delivery agents lies in the recently developedpowerful approaches in phage display and combinatoriallibrary technologies for discovering and screening newcancer-specific peptides [34-37]. Cancer target-specific peptides

Table 1. Examples of colloidal delivery system-based drugs available for cancer treatment.

Drug name Delivery system Mode of action Trade name and company

Doxorubicin citrate Liposome Anthracyclin,DNA intercalator

DaunoXome® – Gilead SciencesMyocet™ – Zeneus Pharma, Elan Corporation

Doxorubicin HCl Liposome Anthracyclin,DNA intercalator

Liposome encapsulated doxorubicin – Neopharm

Doxorubicin HCl Pegylatedliposome

Anthracyclin,DNA intercalator

Doxil® – Tibotec Therapeutics;Caelyx® – Schering-Plough

Lurtotecan Liposome Topoisomerase inhibitor NX211 – OSI Pharmaceuticals

Vincristine Liposome Antimicrotubule agent Marqibo™ – INEX Pharmaceuticals

Carmustine (BCNU) Polymer conjugate Alkylating agents Gliadel® – Guilford Pharmaceuticals

Cytarabine Lipid vesicles Cytosine arabinoside, Antimetabolite

Depocyt® – Skyepharma/Enzon, Pharmacia Coporation, Paladin

Triptorelin pamoate Biodegradable microgranules Gonadotropin inhibitor Trelstar depot® – Debiopharm

Pegylated liposome: Polyethyleneglycol-coated liposome

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

iteit

Tw

ente

on

11/2

0/14

For

pers

onal

use

onl

y.

Page 4: Selective delivery of therapeutic agents for the diagnosis and treatment of cancer

Selective delivery of therapeutic agents for the diagnosis and treatment of cancer

42 Expert Opin. Biol. Ther. (2006) 6(1)

have been used to deliver a variety of therapeutic agents tocancer cells for diagnosis and treatment [38-43]. Peptides havebeen reported to selectively deliver semiconductor quantumdots [44], fluorescence imaging agents, or radioisotopes [43,45]

to cell-specific normal and cancer targets in animals. Recently,experimental molecular diagnostic imaging of oncogenemRNAs has been performed with the help of insulin-likegrowth factor (IGF)1, a receptor-binding peptide in nudemice bearing human IGF1 receptor-overexpressing MCF7breast cancer xenografts. MYC peptide nucleic acid probesconstructed of an IGF1 peptide loop on the C-terminus and a[99mTc]chelator peptide on the N-terminus were visualisedin the tumours using scintigraphic detection technique [46].

5. Selective delivery through nucleotide aptamers

High-affinity aptamers (short DNA or RNA oligonucleotideligands) as targeted therapeutics for the diagnosis and treatmentof cancers are also in development [47-49]. Combinatorialoligonucleotide libraries have been used for the selection ofaptamers that bind to a well-characterised and established

cancer marker selectively and with high affinity [50-53]. Theaptamers in conjugation with appropriate molecules may beuseful for imaging and treatment of cancer [54].

6. Selective delivery by prodrug activation

In order to diminish severe side effects of systemicchemotherapeutic treatment, several prodrug strategies havebeen evolved. Selective delivery of cancer therapeutics is doneby administering a less toxic prodrug form that can beconverted into an active drug at the cancer site. Most of theprodrugs are designed to have a ‘trigger’, ‘linker’ and ‘effector’.The ‘trigger’, following cancer-specific metabolism, modifiesthe ‘linker’, resulting in an activation of the ‘effector’. There areseveral mechanisms potentially exploitable for selective activa-tion. Some utilise unique aspects of tumour physiology (such asselective enzyme expression or hypoxia), whereas others arebased on tumour antigen-specific delivery techniques.

6.1 Targeting endogenous enzymes of cancer cellsIn this approach, systemically administered non-toxic prodrugsare transformed into active drugs at the cancer site as a result of

Table 2. A list of the approved monoclonal antibodies for cancer treatment. Some are already being used for selective delivery of therapeutics in cancer patients and others have a potential for their use as delivery vehicles in future.

mAb name(Approval year)

Target Treatment of Naked/conjugate Trade name Manufacturer

Gemtuzumab ozogamicin(2000)

CD33 Acute myelogenous leukaemia

Calicheamicin, cytotoxic antibiotic

Mylotarg Wyeth, Madison, NJ

Ibritumomab tiuxetan (2002)

CD20 Non-Hodgkin’s lymphoma

Radioisotopes In-111 or Y-90

Zevalin IDEC Pharmaceuticals Corp, San Diego, CA,USA

Iodine I 131 Tositumomab (2003)

CD20 Non-Hodgkin’s lymphoma

Radioisotope I-131 Bexxar® GlaxoSmithKline, Research Triangle Park, NC, USA

Rituximab (1997) CD20 Non-Hodgkin’s lymphoma

Naked* Rituxan® Genentech, Inc., South San Francisco, CA, USA

Trastuzumab (1998) HER-2 Breast cancer Naked* Herceptin® Genentech, Inc., South San Francisco, CA, USA

Alemtuzumab (2001)

CD52 Chronic lymphocytic leukaemia

Naked* Campath® Millennium and ILEX Partners, Cambridge, MA, USA

Cetuximab (2004) EGFR Colorectal cancer Naked* Erbitux™ ImClone Systems, Inc., Branchburg, NJ, USA

Bevacizumab (2004) VEGF Colorectal cancer Naked* Avastin™ Genentech, Inc., South San Francisco, CA, USA

*Antibodies that are approved for cancer treatment and have potential to be used in the future as vehicles for selective delivery of therapeutics in cancer diagnosis and treatment.EGFR: Epidermal growth factor receptor; VEGF: Vascular endothelial growth factor.

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

iteit

Tw

ente

on

11/2

0/14

For

pers

onal

use

onl

y.

Page 5: Selective delivery of therapeutic agents for the diagnosis and treatment of cancer

Shukla & Krag

Expert Opin. Biol. Ther. (2006) 6(1) 43

cancer cell-specific metabolism. Consistently overexpressedendogenous enzymes of tumour cells have been used for theactivation of prodrugs in experimental studies [55]. Clinicaltrials with alkylating indoloquinone EO9 in 38 patients withadvanced non-small cell lung cancer [56], 22 patients withbreast cancer, 26 patients with colon cancer, 20 patients withgastric cancer and 24 patients with pancreatic cancer [57] havebeen reported. However, the results of these clinical studieshave been disappointing and largely attributed to varying levelsof endogenous enzyme activities in tumours [58].

6.2 Targeting secreted enzymes from cancer cellsAnother approach uses prodrugs that are excluded from thecells until cleaved by an enzyme produced and secretedpeferentially by the tumour cells. The peptide–doxorubicinprodrug, L-377202, activated by serine proteaseprostate-specific antigen (PSA) has been shown to selectivelykill prostate tumour cells positive for PSA in an animalmodel [59]. The L-377202 prodrug was ∼ 15 times moreeffective than was doxorubicin at maximally tolerated doses ininhibiting the growth of human prostate cancer tumours innude mice. In a Phase I clinical study in 9 patients withadvanced hormone-resistant prostate cancer, the doxorubicinconjugate of this heptapeptide was well tolerated and found todecrease PSA levels [60].

6.3 Targeting tumour hypoxiaHypoxia is an inherent and potentially exploitable feature ofmost solid tumour types [61]. Hypoxia may occur due toimperfect neovascularisation and compression or obstruction of

blood vessels due to high interstiatial pressures. These featuresare commonly observed during rapid growth of tumours [62].Advances in the chemistry of bioreductive drug activation haveled to the design of hypoxia-selective drug delivery systems.These prodrugs initially undergo one-electron reduction byreductases to give the radical anion, which in normal cells arereoxidised to the parent compound, but in hypoxic tumourcells they are further reduced to more hydrophilic species andtrapped inside. These drugs can be selectively delivered totumours with defined hypoxic fractions rich in the requiredactivating enzymes. Some of the bioreductive prodrugs havenow reached advanced stages of development, including clinicalevaluation; for example, tirapazamine [63-66], AQ4N [67-69] andporfiromycin [70]. The results of these clinical studies were veryencouraging, particularly in cases where bioreductive prodrugswere used in conjunction with radiation and conventionalchemotherapeutic agents. Certain metal-based hypoxic-selec-tive agents for therapy and imaging are under development atpresent [71]. Recently, attempts have been made to targettumour hypoxia for imaging purposes [72], which may help indeveloping tumour profile and treatment decision.

Some examples of the clinical and experimental studies thatutilised tumour enzymes for prodrug therapy are summarisedin Table 3.

6.4 Antibody-directed enzyme prodrug therapyAntibody-directed prodrug therapy (ADEPT) is a 2-stepapproach in which first the antibody–enzyme construct isadministered intravenously. This is composed of an antibodyagainst a tumour-specific target linked to an enzyme that

Table 3. Examples of exploiting tumour enzymes in prodrug cancer therapy in various experimental and clinical studies.

Enzymes Prodrugs Model systems

Endogenous enzymesDT-Diphorase Quinone E09

Tiazofurin

Phase II clinical trial in 92 patients with breast, gastric, pancreatic and colorectal cancer [56]; Multi-centre randomised trial in 38 patients with non-small cell lung cancer [57].Experimental studies [55].

Secreted enzymes

Serine protease PSA L-377202 (DOX–peptide conjugate)

Safety and pharmacokinetics studies in 9 patients with prostate cancer [60]; Experimental studies [59].

Hypoxia-targeted

Cy P450/Cy P450 reductase

NADPH: Cy P450 reductase (CYP3A)

Tirapazamine

AQ4NPorfiromycin

Metal-based prodrugs

Phase I study in 13 patients with lung cancer [65]; 16 patients with oropharyngeal tumours [66]; Phase I/II study in 72 patients with non-small cell lung cancer [63].Experimental studies and clinical trials [67-69].Phase I toxicity trial in 21 patients and a Phase III trial in 34 patients with head and neck squamous cell carcinoma [70].Experimental studies [71].

AQ4n: 1,4-Bis([2-(dimethylamino-N-oxide)ethyl]amino)5,8-dihydroxy- anthracene-9,10-dione), an alkylaminoanthraquinone N-oxide; L-377202: Peptide N-glutaryl-(4-hydroxyprolyl)AlaSer-cyclohexylglycyl-GlnSerLeu-CO2H linked to the aminoglycoside portion of doxorubicin; Porfiromycin: 6-Amino-1,1a,2,8,8a,8b-hexahydro-8-(hydroxymethyl)-8a-methoxy-1,5-dimethyl-azirino[2’,3’:3,4]pyrrolo[1,2-a]indole-4,7-dione, carbamate ester; PSA: Prostate-specific antigen; Quinone E09: Synthetic indoloquinone, structurally related to mytocin C; Tiazofurin: 2-β-D-Ribofuranosyl-4-thiazolecarboxamide; Tirapazamine: 3-Amino-1,2,4-benzotriazine-1,4-dioxide (SR-4233).

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

iteit

Tw

ente

on

11/2

0/14

For

pers

onal

use

onl

y.

Page 6: Selective delivery of therapeutic agents for the diagnosis and treatment of cancer

Selective delivery of therapeutic agents for the diagnosis and treatment of cancer

44 Expert Opin. Biol. Ther. (2006) 6(1)

activates a prodrug. In the second step, after the unboundantibody–enzyme conjugate construct is cleared from thecirculation, a prodrug is administered intravenously. Theprodrug is an anticancer drug that has been rendered lessactive by chemical addition of enzyme-cleavable moieties. Theprodrug is converted to an active form by the tumour-boundantibody–enzyme, which results in local accumulation of theactive form of the anticancer drug [73]. Due to the extracellu-lar site of enzyme action, most of the enzymes used inADEPT studies are hydrolysing enzymes that do not requirecofactors. A variety of enzymes, such as β-lactamase [74-80],cytosine deaminase [81], β-glucuronidase [82-85], carboxypepti-dase A1 [86] and alkaline phosphatase [87-89], and theircounterpart prodrugs have been used in many experimentalstudies. The choice of using non-mammalian enzymeswithout a human homologue helps to achieve high selectivityin ADEPT, but these enzymes raise an immunogenic responsethat can limit duration of the therapy. Table 4 presentsexamples of enzymes and their prodrug substrates used inADEPT clinical trials and experimental studies.

Early ADEPT clinical trials were conducted using theenzyme carboxypeptidase G2 linked to F(ab’)2 fragment ofmurine A5B7 mAb and the benzoic acid mustard prodrugCMDA [90-93]. The infusion of galactosylated second clearingantibody against carboxypeptidase G2 enzyme was done tolower the conjugate levels in the circulation and othernon-tumour tissues before prodrug administration in theseclinical trials. The results were mixed, ranging from no responseto reduction or disappearances of colon tumour deposit andliver metastases. In another Phase I clinical trial, ADEPT wasgiven to 27 patients using A5CP antibody–carboxypeptidase G2conjugate and carbamate prodrug bis-iodo phenol mustard

(ZD2767P) without the treatment with galactosylatedclearing antibody [94]. There were no clinical or radiologicalresponses seen in this study, but 3 patients had stable diseaseat day 56. In the ADEPT clinical trials mentioned above, allthe patients developed antibodies to mouse IgG and enzyme,and cyclosporin treatment was given to suppress the immuneresponse for multiple rounds of therapy. ADEPT faces somevery important challenges related to the large size oftumour-targeting antibody, which include a slow conjugateclearance from blood and normal tissues, limited delivery inpoorly vascularised tumours, and immunogenicity [93,95,96].Studies have shown that certain modifications in fusionproteins could be helpful in reducing some of these problems.Recently, an accelerated clearance of a multifunctionalmannosylated fusion protein from circulation, via hepaticmannose receptors, has been reported in a preclinical study [97].The identification and silencing of epitopes recognised by thehuman immune system have also produced some success inreducing antibody response to mAb conjugates in ADEPT [98].Furthermore, animal studies with xenograft models have shownthat enzyme conjugates of smaller size, in which the ligand unitis a scFv or peptide, may overcome some of the limitationsrelated to antibody molecules [75,84,85,99-101].

6.5 Gene-directed enzyme prodrug therapyGene-directed enzyme prodrug therapy (GDEPT), alsoknown as suicide gene therapy, delivers and expresses the genecoding for an enzyme that is capable of activating a systemi-cally administered prodrug within cancer cells. This can resultin high concentrations of cytotoxic drug at cancer siteswithout harming the rest of the body [102]. In experimentalstudies, genes of several of the enzymes employed for ADEPT

Table 4. Examples of exogenous enzymes used in antibody-directed enzyme prodrug therapy in various experimental and clinical studies.

Enzymes Prodrugs Model systems

Non-mammalian enzymes with no human homologueCarboxypeptidase G2

β-LactamaseCytosine deaminase

Glutamated prodrug: benzoic acidmustard (CMDA)

Bis-iodo phenol mustard (ZD2767P)

Cephalosporin prodrugs5-Fluorocytosine

Clinical trials in 17 patients with colorectal cancer [90,91]; Ten patients with colorectal cancer [92]; Pharmacokinetic study [93]. These used clearing Ab to effectively remove mAb–enzyme before prodrug treatment.Clinical trial in 27 patients, without second clearing Ab [94].Experimental studies [74-80].Experimental studies [81].

Mammalian enzymes, including human enzymesβ-GlucuronidaseCarbxypeptidase A1Alkaline phosphatase

Glucuronide prodrugsα-Peptide prodrugsPhosphate prodrugs

Experimental studies [82-85].Experimental studies [86].Experimental studies [87-89].

α-Peptide prodrugs: For example, methotrexate alpha-peptides; Cephalosporin prodrugs: 7-(4-Carboxybutanamido)cephalosporin conjugates of chemotoxic drugs, such as paclitaxel, doxorubicin, mustard; CMDA: 4-[(2-Chloroethyl) (2-mesyloxyethyl)amino]-benzoyl-L-glutamic acid; Glucuronide prodrugs: For example, N-[4-(daunorubicin or doxorubicin-N-carbonyl(-oxymethyl) phenyl] O-beta-glucuronyl carbamate; Phosphate prodrugs: For example, 4’-Demethylepipodophyllotoxin 9-[4,6-O-(R)-ethylidene-(β)-D-glucopyranoside] with 4’-(dihydrogen phosphate) also called etoposide phosphate, p-[N,N-bis(2-chloroethyl)amino]phenyl phosphate, mitocin phosphate; ZD2767P: [4-[bis(2-iodoethyl)-amino]phenyl oxy carbonyl]-L-glutamic acid.

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

iteit

Tw

ente

on

11/2

0/14

For

pers

onal

use

onl

y.

Page 7: Selective delivery of therapeutic agents for the diagnosis and treatment of cancer

Shukla & Krag

Expert Opin. Biol. Ther. (2006) 6(1) 45

were used for GDEPT [103]. This approach uses both biologi-cal and chemical vector systems available for gene therapy,such as replicable and non-replicable viruses, bacteria andcolloidal systems. When using a viral vector for gene deliveryand expression, GDEPT is sometimes referred to asvirus-directed enzyme prodrug therapy.

Several clinical trials using either adenoviral or retroviraltransduction of herpes simplex virus thymidine kinase(HSV-tk) and ganciclovir prodrug have been conducted incancer patients [104-110]. The GDEPT with HSV-tk has alsobeen reported for positron emission tomography (PET) inglioblastoma patients using prodrug 2’-fluoro-2’-deoxy-1β-D-arabino-furanosyl-5-[124I]iodo-uracil [111]. Several othergenes, such as cytochrome P4502B1 [112], nitroreductase [113]

and cytosine deaminase [114], have been employed for enzymeprodrug therapy trials in cancer patients. Genetically modi-fied, non-pathogenic bacteria [115-119] and allogenic cells [112]

have also been investigated as potential vectors in GDEPT.Different strategies have been adopted to direct these vectorsto cancer sites. These include direct intratumoural inocula-tion or ligand-mediated targeting to cancer-specific antigen.These ligands are chemically conjugated to vectors orexpressed on the coat protein of viral vectors. The use ofbifunctional bridging agents recognising both the virus andthe cancer cell antigen is also common [120]. Viruses aremodified to diminish the binding to their natural receptors onthe cells. The cancer cell-specific ligands, as in other tumour-selective systems, include mAbs, scFvs, peptides or nucleotideaptamers. Another variation of GDEPT that exploits knowntranscriptional differences between normal and tumour cells

to drive selective expression of a prodrug metabolising enzymeis called genetic prodrug activation therapy. It is similar totranscriptional targeting in gene therapy. In this approach, thetrans-response element of a known overexpressed cancer geneis placed upstream of the enzyme gene, resulting in a robustexpression of exogenous enzyme in cancer cells in comparisonto other cells. A clinical trial with cytosine deaminaseexpressed by erbB2-promoter followed by 5-fluorocytosineprodrug treatment has been conducted in breast cancerpatients [114]. Table 5 presents examples of GDEPT clinicaltrials using different enzymes, prodrugs and vectors.

GDEPT has the advantage of using those enzymes that alsorequire endogenous cofactors, but it faces most of thechallenges of gene therapy – right from vector delivery toenzyme expression. Many of the enzymes used in GDEPT actintracellularly by converting prodrugs to active drugs withincells [103]; however, an extracellular effector system comprisedof the secreted form of lysosomal β-glucuronidase andglucuronidated doxorubicin prodrug has also beenreported [121]. In another development, the use of non-invasive19F magnetic resonance spectroscopy and diffusion-weightedMRI as tools to assess gene function and GDEPT efficacy hasbeen demonstrated following administration of a construct ofbifunctional fusion gene between cytosine deaminase anduracil phosphoribosyltransferase [122].

7. External energy-controlled delivery

Some selective delivery strategies involve focusing externalenergy for concentrating or delivering therapeutics at the

Table 5. Examples of clinical studies on gene-directed enzyme prodrug therapy.

Gene Prodrug Vector Study details

HSV-tk

CYP2B1

124I-FIAU

Ifosfamide

Liposome

Allogenic cells

Positron-emission tomography studies with 5 glioblastoma patients in Phase I/II clinical trial [111].Phase I/II clinical trial in 14 pancreatic cancer patients given cells to tumour vessels [112].

Bacterial nitroreductaseHSV-tk

HSV-tkHSV-tkHSV-tk

HSV-tk

HSV-tk

HSV-tk

CB1954

GCV

GCV/VCVVCVGCV

GCV

GCV/VCV

GCV

RD-ADV

RD-ADV

RD-ADVRD-ADVRD-REV

RD-ADV

RD-ADV

RC-REV

Clinical trial in which 8 patients with primary or secondary (colorectal) liver cancer received direct intratumoural inoculation [113].Phase I clinical trial with 18 prostate cancer patients receiving injection directly into the prostate [105].Multi-centre clinical trial in 85 prostate cancer patients [106].Phase I/II clinical trial with 30 prostate cancer patients [107].Phase III multi-centre clinical trial in which 248 glioblastoma multiforme patients received intracerebral injection [110].Clinical trial in 21 malignant pleural mesothelioma receivingpleural injection [108].Clinical trial in 52 prostate cancer patients receiving injection into the prostate [109].Clinical trial in which 48 glioblastoma multiforme patients received intracerebral injection of suspension of retroviral vector-producing cells [104].

Cytosine deaminase

5-Fluorocytosine Directed by erbB2 promoter

Phase I clinical trial using direct intratumoural injection of plasmid construct in 12 breast cancer patients [114].

124I-FIAU: 2’-Fluoro-2’-deoxy-1β-D-arabino-furanosyl-5-[124I]iodo-uracil; Allogenic cells: CYP2B1 enzyme-expressing genetically modified cells; CB1954: 5-(Aziridin-1-yl)-2,4-dinitrobenzamide; CYP2B1: Cytochrome P4502B1; GCV: Ganciclovir prodrug; HSV-tk: Herpes simplex virus thymidine kinase gene; RD-ADV: Replication-deficient adenovirus; RC-REV: Replication-competent retrovirus; RD-REV: Replication-deficient retrovirus; VCV: Valacyclovir prodrug.

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

iteit

Tw

ente

on

11/2

0/14

For

pers

onal

use

onl

y.

Page 8: Selective delivery of therapeutic agents for the diagnosis and treatment of cancer

Selective delivery of therapeutic agents for the diagnosis and treatment of cancer

46 Expert Opin. Biol. Ther. (2006) 6(1)

cancer site. A variety of delivery systems in this category are inthe experimental stage, although some have been used inclinical trials as well.

7.1 Selective delivery through photodynamic therapyPhotodynamic therapy or photochemotherapy destroyscancer cells through the use of a wavelength-specific laser lightin combination with a photosensitising agent. Depending onthe part of the body being treated, the drug is either injectedinto the bloodstream or applied to the skin. The fibre-optic isplaced close to the cancer to deliver the proper amount oflight. After light exposure, the photosensitising agentsproduce radical oxygen species that kill cancer cells. Attemptshave been made to increase the uptake of photosensitisingdrugs by the cancer cells and precise delivery of light to cancersite [123-126]. The fibre-optic can be directed through abronchoscope into the lungs for the treatment of lung cancer,or through an endoscope into the oesophagus for thetreatment of oesophageal cancer [127].

Another modality that uses near-infrared light forlocalised therapeutic thermal ablation and imaging oftumour cells following treatment with gold-coatednanoshells has also been reported [128,129].

7.2 Magnetically targeted deliveryMagnetic-targeted carriers bound to chemotherapeutic orradiological agents in blood circulation can be selectivelyconcentrated at a tumour site using an external constantmagnetic field [130]. Furthermore, thermal activation of aninactive prodrug entrapped in magnetoliposomes using ahigh-frequency magnetic field has been proposed [131].Site-directed thermal ablation of tumour triggered by externalmagnetic field has also been reported [132-135]. The first clini-cal trial using magnetic microspheres loaded with cytotoxicagent was performed in Germany in 14 patients with solidtumours [136]. In a clinical trial with 4 hepatocellularcarcinoma patients, an external magnet was used successfullyto restrict the doxorubicin bound to metallic iron-activatedparticles at the cancer site following MRI-guided transcatheterdelivery to hepatic artery [137].

7.3 Selective delivery through X-ray exposureIn experimental studies with animal models, another class oftargeting methods uses external energy as a trigger forlocalised activation of cytotoxicity. X-rays were used toactivate therapeutic genes driven by a radiation-induciblepromoter of the Egr-1 gene [138]. The Egr–TNF construct wasdelivered through a replication-deficient adenovirus to humantumours growing in nude mice. Exposure of tumours toX-rays resulted in increased TNF-α production and tumourgrowth control without normal tissue damage.

7.4 Radiation-induced selective deliveryRadiation has been used to selectively deliver the drugs tospecific sites such as neoplasms or aberrant blood vessels. It is

based on the fact that blood vessels following exposure toionising radiation express a number of cell adhesion moleculesand receptors, such as intercellular adhesion molecule(ICAM)-1, E-selectin, P-selectin and the β3 integrin, whichcan be used as a target for specific delivery of therapeutics.The experimental studies with animal models havedemonstrated that the ligand-conjugated therapeutics againstthese radiation-induced targets can selectively deliver drugsand imaging agents to tumours following focusedradiation [139,140]. Phase I pharmacokinetic studies onradiation-controlled drug delivery systems have shown thatthe schedule of administration of the therapeutic conjugate inrelation to radiation exposure is crucial [141].

7.5 Ultrasound-guided deliveryThe technique is based on encapsulation of therapeutic agentsin polymeric micelles, passive trapping of the micelles in atumour, and local release of the drug at the target site. The gasesentrapped within lipid coatings make microbubbles sufficientlystable for circulation, and at the same time they can be cavitatedwith focused ultrasound energy for site-specific local delivery ofbioactive materials. It permits using lower concentrations ofdrugs systemically and concentrates the drug at the ultrasound-irradiated site. This technique can be used for both diagnosticand therapeutic applications [142]. There is also a possibility ofattaching microbubbles to site-specific ligands for moleculartargeting. The lipid-coated perfluorocarbon gas-based micro-bubble products are currently in market for diagnostic ultra-sound imaging. Studies in animal tumour models havedemonstrated the selective delivery of chemotherapeutics, genesand imaging agents from acoustically active micelles at thetumour site following exposure to focused ultrasound [143,144].

8. Organ-selective delivery

Attempts have also been made for organ-specific delivery ofcancer therapeutics. A methotrexate–bisphosphonate conju-gate containing a peptide bond has been found to possess > 5times more antineoplastic activity against osteosarcoma inexperimental animal models than methotrexate alone [145].This approach is based on the high affinity of P-C-P portionof the bisphosphonate to hydroxyapatite, which is a majorbone constituent. These developments which are being testedfor bone-selective delivery of a variety of agents [146] areimportant because drug delivery to bone is limited by itsunique anatomical features. Organ-specific pharmaceuticalapproaches have also been used for the colon and liver [147,148].

In addition to tumour targeting, developing successfultechnology for a selective delivery requires an appropriateformulation that helps to avoid sequestration of the targetingmolecules by the reticuloendothelium system, to reduce theclearance of small drugs, to provide protection of active agentsfrom degradation, to overcome the biological barriers thatprevent it reaching the target, and to tackle the augmentedosmotic pressure state in cancer lesions.

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

iteit

Tw

ente

on

11/2

0/14

For

pers

onal

use

onl

y.

Page 9: Selective delivery of therapeutic agents for the diagnosis and treatment of cancer

Shukla & Krag

Expert Opin. Biol. Ther. (2006) 6(1) 47

9. Expert opinion and conclusion

Despite a number of advances in the past few decades,systemic cancer therapy is hampered by problems of severeunwanted side effects and the development of drug resistance.Research activity aimed towards achieving specific andtargeted delivery of cancer therapeutics has expanded tremen-dously in the last decade with new methods of directing drugsto tumours, as well as drugs with new mechanisms of action.At this moment, it is not clear whether some of the cancertargeting therapeutic strategies presented in this review, inspite of showing encouraging results in animal models, will besuccessful in the clinic. The success in this field will rely onthe vector system and delivery strategy, exploration of newtargets and their high-affinity ligands, better understanding ofthe biology of cancer, and the invention of more clinicallyrelevant models for human cancer research. Ultimately, muchof what we will learn about tumour-selective delivery in thenext few years will depend on how rapidly clinical-gradereagents will become available, and on the determination ofboth industrial and academic groups to perform translationalclinical research on these strategies.

For the success of ligand-mediated selective deliverystrategies, it is essential to develop high-affinity ligands againstvalidated cancer targets. The discovery and validation oftumour-associated markers as well as corresponding ligandsremain an important challenge for the development oftechnology for the biomarker-targeted delivery of therapeuticagents for the treatment and imaging of solidtumours [3,4,149,150]. Although mAbs offer excellent specificityand affinity, and their use in the haematological malignancieshas shown great promise, poor barrier penetration is animportant issue in solid tumour treatment. It has beenrecognised that only 0.001 – 0.01% of intravenously adminis-tered mAbs reach their parenchymal targets in vivo [151].Targeting cancer-specific vascular targets for selective deliveryof mAb-conjugated therapeutics to tumour sites shouldminimise the problems associated with physiological barriersto the delivery of macromolecules to parenchymal targets [41].This strategy could be especially useful in ADEPT because ofa bystander effect on cancer cells owing to enzymatic genera-tion of small diffusible active drug molecules at tumour site.Advances in genetic engineering of antibodies have generatedbispecific antibodies that have one targeting arm to recognisetumour antigens and another to bind effectors. Pretargetingwith such antibodies allows time for tumour-target bindingand clearance from circulation, before toxic effectors areadministered [21]. The technology also led to the developmentof catalytic antibodies or abzymes that may find use inADEPT [152,153]. Clinical trials with targeted delivery of drugshave shown promise with prodrug therapy. Two amplifica-tions steps are obtained with prodrug therapy. The first isthat the activating ligand–enzyme conjugate is present athigher concentration in the tumour target. The secondamplification step is the step up achieved by enzyme catalysis

of one mAb–enzyme conjugate molecule activating manyprodrug molecules to an active cytotoxic state.

In the near future, antibody fragments and peptide ligandsgenerated by molecular methods may lead to improvementsover antibodies generated in an animal. Using phage displaytechnology, rapid identification of such ligands againstcancer-specific targets should facilitate the development oftumour-specific reagents and may even lead to individualisedtherapy. The economical bulk production of such proteins inbacterial hosts is an added advantage over the difficulties ofproducing large quantities of monoclonal antibodies requiredfor clinical use. Recently, we screened a phage-displayedβ-lactamase scaffold library in which an outer loop of theenzyme was randomised, and selected a number ofhigh-affinity cancer-binding clones (under preparation).These targeted enzymes, which bring together in onemolecule a tumour-binding module and an effector module,are much smaller than an antibody–enzyme construct. Severalphage-derived products are in the clinical pipeline and somehave been approved by the US Food and Drug Administrationfor human use [3,154].

Nucleotide aptamers are also very promising candidates astumour-targeting ligands because of their high affinity andease in selection against numerous targets. The new genera-tion of aptamers or ‘spiegelmers’ are nuclease-resistant, whichmakes them more stable under biological conditions [155,156].Small molecular weight molecules are an attractive alternativeto peptides and nucleotides as tumour-specific deliveryvehicles. They have good penetrative characteristics, can bereadily conjugated to effectors and can have a high affinity forantigens expressed on tumour cell surface. Such small organiccompounds are expected to be non-immunogenic and shoulddisplay an improved tumour penetration compared withantibodies. Folic acid is an example of a small moleculeligand that binds with high-affinity to membrane-boundfolate receptors known to be overexpressed on a variety ofhuman cancers. Folic acid has been reported to selectivelydeliver a wide array of imaging and toxic agents to suchcancers [157]. The developments in the generation of syntheticcombinatorial libraries of a class of organic chemicals,commonly referred to as ‘small molecules’, and theirhigh-throughput screening have paved the way for develop-ing this new class of cancer target-specific ligands for selectivedelivery in cancer imaging and treatment. It is expected toprovide high affinity ligands for antigens with cavities (e.g.,enzymes), but isolation of high-affinity ligands for flatproteins could be a challenge. Improvements in the technolo-gies for isolating high-affinity and high-specificity smallmolecules will allow cancer-targeting experiments to becarried out with classes of small molecules having differentpharmacokinetic properties [158-160].

Although several GDEPT clinical trials have shown someencouraging results, the strategies that require a gene transferstep in the selective delivery of therapeutics have to deal withall the challenges that gene therapy faces today. These include

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

iteit

Tw

ente

on

11/2

0/14

For

pers

onal

use

onl

y.

Page 10: Selective delivery of therapeutic agents for the diagnosis and treatment of cancer

Selective delivery of therapeutic agents for the diagnosis and treatment of cancer

48 Expert Opin. Biol. Ther. (2006) 6(1)

poor gene transfer, degradation and immune attack of vectors,control and regulation of gene expression, environmentalsafety and, above all, patient toxicity [161]. According to TheJournal of Gene Medicine website ‘Gene Therapy ClinicalTrials Worldwide’ [201], 7.6% of the trials are in the field ofsuicide gene therapy. Vehicles such as retroviruses, adeno-viruses, and non-viral systems adapted for clinical trialsemploying different strategies of selective delivery haveproduced mixed results. The future of such strategies will behighly dependent on the development of safe and effectivevectors for gene transfer.

The use of external energy-guided delivery technologiesdepends on the exposure modality and the location oftumours; therefore, their application is limited only tospecific situations. For example, photodynamic therapy issuitable only for tumours that can be exposed to focusedlight. However, for obstruction of the gut or airway byadvanced tumours, such directed treatment, albeit limitedand non-systemic, may provide substantial benefit. In thenear future, the external energy-guided delivery systems areexpected to be more often coupled with other molecularlytargeted delivery systems, such as those employinghigh-affinity ligands.

The bioavailability of the selectively delivered agents at thedesired site depends on several characteristics, includingmolecular size; hydrophilicity; charge; stability; rate of elimi-nation through degradation, secretion and nonspecificbinding; and tissue specific factors such as interstitial pressuresand vascular permeability. Advances in the field of nanotech-nology have helped create a new generation of nanovectors.These not only avoid biobarriers, but also allow placement ofdesired targeting moieties on the surface of the particles, aswell as loading the particles with different imaging and/ortherapeutic agents [7]. Such nanovectors may significantlyincrease the number of therapeutic molecules that can be

delivered per tumour target molecule and can allow sustainedrelease of the therapy over time. Recently, the development ofa delivery vehicle ‘nanocell’ for the sequential delivery of twodrugs makes it possible to target distinct tumour compart-ments or multiple signalling pathways [9]. By systematicallycombining preferred therapeutic agents and cancer-targetingmoieties, nanotechnology platform may help to generatenovel, personalised agents for selective delivery in the diagnosisand treatment of cancer.

The success of biomarker-targeted delivery technology willdepend on the identification of suitable markers of neoplasticdisease and understanding target evolution over time. Deploy-ment of these markers in advanced drug delivery systems willfacilitate diagnostic screening and effective treatment ofcancer. Advanced imaging modalities in combination withcancer target-specific probes provide a powerful non-invasivediagnostic tool. Molecular diagnostic imaging is an emergingtechnology that may distinguish patient populations thatwould respond to targeted therapy from non-responders. Theconvergence of genomic technologies and the development ofdrugs designed against specific molecular targets are expectedto provide many opportunities for cancer diagnosis andtreatment in near future. The development of suitabletargeted delivery systems for these new generations of drugsand their successful translation into clinical practice is one ofthe most promising but significant challenges in the field ofoncology today.

Acknowledgements

The work was supported by The National Cancer Institute(R01 CA080790 & R01 CA112091) and US Army MedicalResearch (W81XWH0510237), and in part by the VermontCancer Center core grant (PHS CA22435) and SD IrelandCancer Research fund.

BibliographyPapers of special note have been highlighted as either of interest (•) or of considerable interest (••) to readers.

1. KIRSNER KM: Cancer: new therapies and new approaches to recurring problems. AANA J. (2003) 71(1):55-62.

2. EGGERMONT AM, BRUNSTEIN F, GRUNHAGEN D, TEN HAGEN TL: Regional treatment of metastasis: role of regional perfusion. State of the art isolated limb perfusion for limb salvage. Ann. Oncol. (2004) 15(Suppl. 4):107-112.

3. ALLEN TM: Ligand-targeted therapeutics in anticancer therapy. Nat. Rev. Cancer (2002) 2(10):750-763.

•• A review on ligand-mediated delivery.

4. PEGRAM MD, PIETRAS R, BAJAMONDE A, KLEIN P, FYFE G: Targeted therapy: wave of the future. J. Clin. Oncol. (2005) 23(8):1776-1781.

5. ROSKOSKI R JR: The ErbB/HER receptor protein-tyrosine kinases and cancer. Biochem. Biophys. Res. Commun. (2004) 319(1):1-11.

6. KIM CK, LIM SJ: Recent progress in drug delivery systems for anticancer agents. Arch. Pharm. Res. (2002) 25(3):229-239.

7. FERRARI M: Cancer nanotechnology: opportunities and challenges. Nat. Rev. Cancer (2005) 5(3):161-171.

•• A review on the recent developments in nanovectors for delivery in cancer.

8. GAO X, CUI Y, LEVENSON RM, CHUNG LW, NIE S: In vivo cancer

targeting and imaging with semiconductor quantum dots. Nat. Biotechnol. (2004) 22(8):969-976.

•• In vivo imaging of tumours using quantum dots.

9. SENGUPTA S, EAVARONE D, CAPILA I et al.: Temporal targeting of tumour cells and neovasculature with a nanoscale delivery system. Nature (2005) 436(7050):568-572.

•• Development of nanovectors for sequential delivery of two drugs.

10. DEARDEN C: Monoclonal antibody therapy of haematological malignancies. BioDrugs (2002) 16(4):283-301.

11. FLYNN AA, PEDLEY RB, GREEN AJ et al.: Antibody and radionuclide characteristics and the enhancement of the effectiveness of radioimmunotherapy by selective dose delivery to radiosensitive areas

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

iteit

Tw

ente

on

11/2

0/14

For

pers

onal

use

onl

y.

Page 11: Selective delivery of therapeutic agents for the diagnosis and treatment of cancer

Shukla & Krag

Expert Opin. Biol. Ther. (2006) 6(1) 49

of tumour. Int. J. Radiat. Biol. (2002) 78(5):407-415.

12. MACDONALD GC, GLOVER N: Effective tumor targeting: strategies for the delivery of Armed Antibodies. Curr. Opin. Drug Disc. Dev. (2005) 8(2):177-183.

• A review on the use of mAbs for selective delivery in cancer.

13. MARKS AJ, COOPER MS, ANDERSON RJ et al.: Selective apoptotic killing of malignant hemopoietic cells by antibody-targeted delivery of an amphipathic peptide. Cancer Res. (2005) 65(6):2373-2377.

14. DAMLE NK: Tumour-targeted chemotherapy with immunoconjugates of calicheamicin. Expert Opin. Biol. Ther. (2004) 4(9):1445-1452.

15. BODEY B: Genetically engineered antibodies for direct antineoplastic treatment and systematic delivery of various therapeutic agents to cancer cells. Expert Opin. Biol. Ther. (2001) 1(4):603-617.

• A review on the use of mAbs for selective delivery in cancer.

16. EBBINGHAUS C, RONCA R, KASPAR M et al.: Engineered vascular-targeting antibody-interferon-gamma fusion protein for cancer therapy. Int. J. Cancer (2005) 116(2):304-313.

17. CHESTER K, PEDLEY B, TOLNER B et al.: Engineering antibodies for clinical applications in cancer. Tumour Biol. (2004) 25(1-2):91-98.

18. LI KC, GUCCIONE S, BEDNARSKI MD: Combined vascular targeted imaging and therapy: a paradigm for personalized treatment. J. Cell. Biochem. Suppl. (2002) 39:65-71.

• Selective delivery through molecular targeting for cancer imaging and treatment.

19. GUCCIONE S, LI KC, BEDNARSKI MD: Molecular imaging and therapy directed at the neovasculature in pathologies. How imaging can be incorporated into vascular-targeted delivery systems to generate active therapeutic agents. IEEE Eng. Med. Biol. Mag. (2004) 23(5):50-56.

20. SHAHBAZI-GAHROUEI D, WILLIAMS M, RIZVI S, ALLEN BJ: In vivo studies of Gd-DTPA-monoclonal antibody and gd-porphyrins: potential magnetic resonance imaging contrast agents for melanoma. J. Magn. Reson. Imaging (2001) 14(2):169-174.

21. CHANG CH, SHARKEY RM, ROSSI EA et al.: Molecular advances in pretargeting radioimunotherapy with bispecific antibodies. Mol. Cancer Ther. (2002) 1(7):553-563.

• Use of bispecific mAbs in pretargeting radioimmunotherapy.

22. SANTIMARIA M, MOSCATELLI G, VIALE GL et al.: Immunoscintigraphic detection of the ED-B domain of fibronectin, a marker of angiogenesis, in patients with cancer. Clin. Cancer Res. (2003) 9(2):571-579.

23. LOO C, LOWERY A, HALAS N, WEST J, DREZEK R: Immunotargeted nanoshells for integrated cancer imaging and therapy. Nano Lett. (2005) 5(4):709-711.

• Use of cancer-targeted nanoshells for imaging.

24. GOLDENBERG DM: Targeted therapy of cancer with radiolabeled antibodies. J. Nucl. Med. (2002) 43(5):693-713.

25. QU Z, GRIFFITHS GL, WEGENER WA et al.: Development of humanized antibodies as cancer therapeutics. Methods (2005) 36(1):84-95.

26. NIV R, COHEN CJ, DENKBERG G, SEGAL D, REITER Y: Antibody engineering for targeted therapy of cancer: recombinant Fv-immunotoxins. Curr. Pharm. Biotechnol. (2001) 2(1):19-46.

27. SUZUKI M, TAKAYANAGI A, SHIMIZU N: Targeted gene delivery using humanized single-chain antibody with negatively charged oligopeptide tail. Cancer Sci. (2004) 95(5):424-429.

28. CHESTER KA, BHATIA J, BOXER G et al.: Clinical applications of phage-derived sFvs and sFv fusion proteins. Dis. Markers (2000) 16(1-2):53-62.

29. AZZAZY HM, HIGHSMITH WE JR: Phage display technology: clinical applications and recent innovations. Clin. Biochem. (2002) 35(6):425-445.

• One of the reviews on the developments in phage display technology and their clinical applications.

30. HOOGENBOOM HR: Overview of antibody phage-display technology and its applications. Methods Mol. Biol. (2002) 178:1-37.

31. AINA OH, SROKA TC, CHEN ML, LAM KS: Therapeutic cancer targeting peptides. Biopolymers (2002) 66(3):184-199.

32. JANIN YL: Peptides with anticancer use or potential. Amino Acids (2003) 25(1):1-40.

33. BORGHOUTS C, KUNZ C, GRONER B: Peptide aptamers: recent developments for cancer therapy. Expert Opin. Biol. Ther. (2005) 5(6):783-797.

34. ROMANOV VI: Phage display selection and evaluation of cancer drug targets. Curr. Cancer Drug Targets (2003) 3(2):119-129.

35. NILSSON F, TARLI L, VITI F, NERI D: The use of phage display for the development of tumour targeting agents. Adv. Drug Deliv. Rev. (2000) 43(2-3):165-196.

36. SHUKLA GS, KRAG DN: Phage display selection for cell-specific ligands: development of a screening procedure suitable for small tumor specimens. J. Drug Target. (2005) 13(1):7-18.

37. SHUKLA GS, KRAG DN: Selection of tumor-targeting agents on freshly excised human breast tumors using a phage display library. Oncol. Rep. (2005) 13(4):757-764.

•• First report on using fresh human tumours for phage display selection of cancer-specific ligands.

38. PASQUALINI R, RUOSLAHTI E: Organ targeting in vivo using phage display peptide libraries. Nature (1996) 380(6572):364-366.

•• Targeted delivery of chemotherapeutic agents using cancer-specific peptide ligands.

39. CHEN X, PLASENCIA C, HOU Y, NEAMATI N: Synthesis and biological evaluation of dimeric RGD peptide-paclitaxel conjugate as a model for integrin-targeted drug delivery. J. Med. Chem. (2005) 48(4):1098-1106.

40. MOFFATT S, WIEHLE S, CRISTIANO RJ: Tumor-specific gene delivery mediated by a novel peptide-polyethylenimine-DNA polyplex targeting aminopeptidase N/CD13. Hum. Gene Ther. (2005) 16(1):57-67.

41. KOLONIN M, PASQUALINI R, ARAP W: Molecular addresses in blood vessels as targets for therapy. Curr. Opin. Chem. Biol. (2001) 5(3):308-313.

42. GOODWIN DA, MEARES CF: Pretargeted peptide imaging and therapy. Cancer Biother. Radiopharm. (1999) 14(3):145-152.

43. BUCHSBAUM DJ: Imaging and therapy of tumors induced to express somatostatin receptor by gene transfer using radiolabeled peptides and single chain antibody

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

iteit

Tw

ente

on

11/2

0/14

For

pers

onal

use

onl

y.

Page 12: Selective delivery of therapeutic agents for the diagnosis and treatment of cancer

Selective delivery of therapeutic agents for the diagnosis and treatment of cancer

50 Expert Opin. Biol. Ther. (2006) 6(1)

constructs. Semin. Nucl. Med. (2004) 34(1):32-46.

44. AKERMAN ME, CHAN WC, LAAKKONEN P, BHATIA SN, RUOSLAHTI E: Nanocrystal targeting in vivo. Proc. Natl. Acad. Sci. USA (2002) 99(20):12617-12621.

• Experimental studies on the peptide-mediated delivery of quantum dots for cancer imaging.

45. BUCHSBAUM DJ, CHAUDHURI TR, YAMAMOTO M, ZINN KR: Gene expression imaging with radiolabeled peptides. Ann. Nucl. Med. (2004) 18(4):275-283.

46. TIAN X, ARUVA MR, QIN W et al.: Noninvasive molecular imaging of MYC mRNA expression in human breast cancer xenografts with a [99mTc]peptide-peptide nucleic acid-peptide chimera. Bioconjug. Chem. (2005) 16(1):70-79.

47. FAROKHZAD OC, JON S, KHADEMHOSSEINI A, TRAN TN, LAVAN DA, LANGER R: Nanoparticle-aptamer bioconjugates: a new approach for targeting prostate cancer cells. Cancer Res. (2004) 64(21):7668-7672.

48. NIMJEE SM, RUSCONI CP, SULLENGER BA: Aptamers: an emerging class of therapeutics. Annu. Rev. Med. (2005) 56:555-583.

• One of the recent reviews on aptamers.

49. CERCHIA L, HAMM J, LIBRI D, TAVITIAN B, DE FRANCISCIS V: Nucleic acid aptamers in cancer medicine. FEBS Lett. (2002) 528(1-3):12-16.

50. BLANK M, WEINSCHENK T, PRIEMER M, SCHLUESENER H: Systematic evolution of a DNA aptamer binding to rat brain tumor microvessels. selective targeting of endothelial regulatory protein pigpen. J. Biol. Chem. (2001) 276(19):16464-16468.

51. SURUGIU-WARNMARK I, WARNMARK A, TORESSON G, GUSTAFSSON JA, BULOW L: Selection of DNA aptamers against rat liver X receptors. Biochem. Biophys. Res. Commun. (2005) 332(2):512-517.

52. ELLINGTON AD, SZOSTAK JW: Selection in vitro of single-stranded DNA molecules that fold into specific ligand-binding structures. Nature (1992) 355(6363):850-852.

•• Selection of target-specific aptamers.

53. SCHMIDT KS, BORKOWSKI S, KURRECK J et al.: Application of locked

nucleic acids to improve aptamer in vivo stability and targeting function. Nucleic Acids Res. (2004) 32(19):5757-5765.

54. BOISGARD R, KUHNAST B, VONHOFF S et al.: In vivo biodistribution and pharmacokinetics of 18F-labelled Spiegelmers: a new class of oligonucleotidic radiopharmaceuticals. Eur. J. Nucl. Med. Mol. Imaging (2005) 32(4):470-477.

55. FITZSIMMONS SA, WORKMAN P, GREVER M, PAULL K, CAMALIER R, LEWIS AD: Reductase enzyme expression across the National Cancer Institute Tumor cell line panel: correlation with sensitivity to mitomycin C and EO9. J. Natl. Cancer Inst. (1996) 88(5):259-269.

56. DIRIX LY, TONNESEN F, CASSIDY J et al.: EO9 Phase II study in advanced breast, gastric, pancreatic and colorectal carcinoma by the EORTC Early Clinical Studies Group. Eur. J. Cancer (1996) 32A(11):2019-2022.

• Clinical trial targeting endogenous enzymes in enzyme prodrug therapy.

57. PAVLIDIS N, HANAUSKE AR, GAMUCCI T et al.: A randomized Phase II study with two schedules of the novel indoloquinone EO9 in non-small-cell lung cancer: a study of the EORTC Early Clinical Studies Group (ECSG). Ann. Oncol. (1996) 7(5):529-531.

•• Clinical trial targeting endogenous tumour enzymes in prodrug therapy.

58. CUMMINGS J, SPANSWICK VJ, GARDINER J, RITCHIE A, SMYTH JF: Pharmacological and biochemical determinants of the antitumour activity of the indoloquinone EO9. Biochem. Pharmacol. (1998) 55(3):253-260.

59. DEFEO-JONES D, GARSKY VM, WONG BK et al.: A peptide-doxorubicin ‘prodrug’ activated by prostate-specific antigen selectively kills prostate tumor cells positive for prostate-specific antigen in vivo. Nat. Med. (2000) 6(11):1248-1252.

60. DIPAOLA RS, RINEHART J, NEMUNAITIS J et al.: Characterization of a novel prostate-specific antigen-activated peptide-doxorubicin conjugate in patients with prostate cancer. J. Clin. Oncol. (2002) 20(7):1874-1879.

•• Clinical trial targeting tumour-secreted enzymes in prodrug therapy.

61. BROWN JM: The hypoxic cell: a target for selective cancer therapy-eighteenth Bruce F. Cain Memorial Award lecture. Cancer Res. (1999) 59(23):5863-5870.

62. HARRIS AL: Hypoxia-a key regulatory factor in tumour growth. Nat. Rev. Cancer (2002) 2(1):38-47.

63. GATINEAU M, RIXE O, CHEVALIER TL: Tirapazamine with cisplatin and vinorelbine in patients with advanced non-small-cell lung cancer: a Phase I/II study. Clin. Lung Cancer (2005) 6(5):293-298.

64. COWEN RL, WILLIAMS KJ, CHINJE EC et al.: Hypoxia targeted gene therapy to increase the efficacy of tirapazamine as an adjuvant to radiotherapy: reversing tumor radioresistance and effecting cure. Cancer Res. (2004) 64(4):1396-1402.

65. JOHNSON CA, KILPATRICK D, VON ROEMELING R et al.: Phase I trial of tirapazamine in combination with cisplatin in a single dose every 3 weeks in patients with solid tumors. J. Clin. Oncol. (1997) 15(2):773-780.

66. RISCHIN D, PETERS L, HICKS R et al.: Phase I trial of concurrent tirapazamine, cisplatin, and radiotherapy in patients with advanced head and neck cancer. J. Clin. Oncol. (2001) 19(2):535-542.

•• Clinical study on a hypoxia-targeted drug in conjunction with conventional chemo- and radiotherapy.

67. PATTERSON LH: Bioreductively activated antitumor N-oxides: the case of AQ4N, a unique approach to hypoxia-activated cancer chemotherapy. Drug Metab. Rev. (2002) 34(3):581-592.

68. PATTERSON LH, MCKEOWN SR, RUPARELIA K et al.: Enhancement of chemotherapy and radiotherapy of murine tumours by AQ4N, a bioreductively activated anti-tumour agent. Br. J. Cancer (2000) 82(12):1984-1990.

69. FRIERY OP, GALLAGHER R, MURRAY MM et al.: Enhancement of the anti-tumour effect of cyclophosphamide by the bioreductive drugs AQ4N and tirapazamine. Br. J. Cancer (2000) 82(8):1469-1473.

70. HAFFTY BG, SON YH, WILSON LD et al.: Bioreductive alkylating agent porfiromycin in combination with radiation therapy for the management of squamous cell carcinoma of the head and neck. Radiat. Oncol. Invest. (1997) 5(5):235-245.

71. BLOWER PJ, DILWORTH JR, MAURER RI, MULLEN GD, REYNOLDS CA, ZHENG Y: Towards new transition metal-based hypoxic selective

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

iteit

Tw

ente

on

11/2

0/14

For

pers

onal

use

onl

y.

Page 13: Selective delivery of therapeutic agents for the diagnosis and treatment of cancer

Shukla & Krag

Expert Opin. Biol. Ther. (2006) 6(1) 51

agents for therapy and imaging. J. Inorg. Biochem. (2001) 85(1):15-22.

72. RAJENDRAN JG, KROHN KA: Imaging hypoxia and angiogenesis in tumors. Radiol. Clin. North Am. (2005) 43(1):169-187.

73. BAGSHAWE KD, SHARMA SK, BEGENT RH: Antibody-directed enzyme prodrug therapy (ADEPT) for cancer. Expert Opin. Biol. Ther. (2004) 4(11):1777-1789.

• One of the reviews on ADEPT.

74. VRUDHULA VM, SVENSSON HP, KENNEDY KA, SENTER PD, WALLACE PM: Antitumor activities of a cephalosporin prodrug in combination with monoclonal antibody-beta-lactamase conjugates. Bioconjug. Chem. (1993) 4(5):334-340.

75. RODRIGUES ML, PRESTA LG, KOTTS CE et al.: Development of a humanized disulfide-stabilized anti-p185HER2 Fv-beta-lactamase fusion protein for activation of a cephalosporin doxorubicin prodrug. Cancer Res. (1995) 55(1):63-70.

•• Use of a single-chain antibody fusion protein in enzyme prodrug therapy.

76. KERR DE, SCHREIBER GJ, VRUDHULA VM et al.: Regressions and cures of melanoma xenografts following treatment with monoclonal antibody beta-lactamase conjugates in combination with anticancer prodrugs. Cancer Res. (1995) 55(16):3558-3563.

77. MEYER DL, LAW KL, PAYNE JK et al.: Site-specific prodrug activation by antibody-beta-lactamase conjugates: preclinical investigation of the efficacy and toxicity of doxorubicin delivered by antibody directed catalysis. Bioconjug. Chem. (1995) 6(4):440-446.

78. SVENSSON HP, FRANK IS, BERRY KK, SENTER PD: Therapeutic effects of monoclonal antibody-beta-lactamase conjugates in combination with a nitrogen mustard anticancer prodrug in models of human renal cell carcinoma. J. Med. Chem. (1998) 41(9):1507-1512.

79. SVENSSON HP, VRUDHULA VM, EMSWILER JE et al.: In vitro and in vivo activities of a doxorubicin prodrug in combination with monoclonal antibody beta-lactamase conjugates. Cancer Res. (1995) 55(11):2357-2365.

80. VRUDHULA VM, KERR DE, SIEMERS NO, DUBOWCHIK GM, SENTER PD: Cephalosporin prodrugs of

paclitaxel for immunologically specific activation by L-49-sFv-beta-lactamase fusion protein. Bioorg. Med. Chem. Lett. (2003) 13(3):539-542.

81. WALLACE PM, MACMASTER JF, SMITH VF, KERR DE, SENTER PD, COSAND WL: Intratumoral generation of 5-fluorouracil mediated by an antibody-cytosine deaminase conjugate in combination with 5-fluorocytosine. Cancer Res. (1994) 54(10):2719-2723.

82. HOUBA PH, BOVEN E, ERKELENS CA et al.: The efficacy of the anthracycline prodrug daunorubicin-GA3 in human ovarian cancer xenografts. Br. J. Cancer (1998) 78(12):1600-1606.

83. BIELA BH, KHAWLI LA, HU P, EPSTEIN AL: Chimeric TNT-3/human beta-glucuronidase fusion proteins for antibody-directed enzyme prodrug therapy (ADEPT). Cancer Biother. Radiopharm. (2003) 18(3):339-353.

84. HAISMA HJ, SERNEE MF, HOOIJBERG E et al.: Construction and characterization of a fusion protein of single-chain anti-CD20 antibody and human beta-glucuronidase for antibody-directed enzyme prodrug therapy. Blood (1998) 92(1):184-190.

85. DE GRAAF M, BOVEN E, OOSTERHOFF D et al.: A fully human anti-Ep-CAM scFv-beta-glucuronidase fusion protein for selective chemotherapy with a glucuronide prodrug. Br. J. Cancer (2002) 86(5):811-818.

86. SMITH GK, BANKS S, BLUMENKOPF TA et al.: Toward antibody-directed enzyme prodrug therapy with the T268G mutant of human carboxypeptidase A1 and novel in vivo stable prodrugs of methotrexate. J. Biol. Chem. (1997) 272(25):15804-15816.

• Use of a mutant human enzyme for ADEPT.

87. WALLACE PM, SENTER PD: In vitro and in vivo activities of monoclonal antibody-alkaline phosphatase conjugates in combination with phenol mustard phosphate. Bioconjug. Chem. (1991) 2(5):349-352.

88. SENTER PD, SAULNIER MG, SCHREIBER GJ et al.: Anti-tumor effects of antibody-alkaline phosphatase conjugates in combination with etoposide phosphate. Proc. Natl. Acad. Sci. USA (1988) 85(13):4842-4846.

• First proof of principle of ADEPT.

89. SENTER PD, SCHREIBER GJ, HIRSCHBERG DL, ASHE SA, HELLSTROM KE, HELLSTROM I: Enhancement of the in vitro and in vivo antitumor activities of phosphorylated mitomycin C and etoposide derivatives by monoclonal antibody-alkaline phosphatase conjugates. Cancer Res. (1989) 49(21):5789-5792.

90. BAGSHAWE KD, SHARMA SK, SPRINGER CJ et al.: Antibody directed enzyme prodrug therapy (ADEPT): clinical report. Dis. Markers (1991) 9(3-4):233-238.

•• First clinical report on ADEPT.

91. BAGSHAWE KD, SHARMA SK, SPRINGER CJ, ANTONIW P: Antibody-directed enzyme prodrug therapy: a pilot scale clinical trial. Tumor Targeting (1995) 1(1):17-29.

92. NAPIER MP, SHARMA SK, SPRINGER CJ et al.: Antibody-directed enzyme prodrug therapy: efficacy and mechanism of action in colorectal carcinoma. Clin. Cancer Res. (2000) 6(3):765-772.

93. MARTIN J, STRIBBLING SM, POON GK et al.: Antibody-directed enzyme prodrug therapy: pharmacokinetics and plasma levels of prodrug and drug in a Phase I clinical trial. Cancer Chemother. Pharmacol. (1997) 40(3):189-201.

94. FRANCIS RJ, SHARMA SK, SPRINGER C et al.: A Phase I trial of antibody directed enzyme prodrug therapy (ADEPT) in patients with advanced colorectal carcinoma or other CEA producing tumours. Br. J. Cancer (2002) 87(6):600-607.

•• A clinical report on ADEPT using bis-iodo phenol mustard prodrug.

95. DENNY WA, WILSON WR: The design of selectively-activated anti-cancer prodrugs for use in antibody-directed and gene-directed enzyme-prodrug therapies. J. Pharm. Pharmacol. (1998) 50(4):387-394.

96. MASON DW, WILLIAMS AF: The kinetics of antibody binding to membrane antigens in solution and at the cell surface. Biochem. J. (1980) 187(1):1-20.

97. SHARMA SK, PEDLEY RB, BHATIA J et al.: Sustained tumor regression of human colorectal cancer xenografts using a multifunctional mannosylated fusion protein in antibody-directed enzyme prodrug therapy. Clin. Cancer Res. (2005) 11(2 Pt 1):814-825.

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

iteit

Tw

ente

on

11/2

0/14

For

pers

onal

use

onl

y.

Page 14: Selective delivery of therapeutic agents for the diagnosis and treatment of cancer

Selective delivery of therapeutic agents for the diagnosis and treatment of cancer

52 Expert Opin. Biol. Ther. (2006) 6(1)

98. MAYER A, SHARMA SK, TOLNER B et al.: Modifying an immunogenic epitope on a therapeutic protein: a step towards an improved system for antibody-directed enzyme prodrug therapy (ADEPT). Br. J. Cancer (2004) 90(12):2402-2410.

99. BHATIA J, SHARMA SK, CHESTER KA et al.: Catalytic activity of an in vivo tumor targeted anti-CEA scFv::carboxypeptidase G2 fusion protein. Int. J. Cancer (2000) 85(4):571-577.

100. MICHAEL NP, CHESTER KA, MELTON RG et al.: In vitro and in vivo characterisation of a recombinant carboxypeptidase G2::anti-CEA scFv fusion protein. Immunotechnology (1996) 2(1):47-57.

101. WONG BK, DEFEO-JONES D, JONES RE et al.: PSA-specific and non-PSA-specific conversion of a PSA-targeted peptide conjugate of doxorubicin to its active metabolites. Drug Metab. Dispos. (2001) 29(3):313-318.

102. DACHS GU, TUPPER J, TOZER GM: From bench to bedside for gene-directed enzyme prodrug therapy of cancer. Anticancer. Drugs (2005) 16(4):349-359.

103. XU G, MCLEOD HL: Strategies for enzyme/prodrug cancer therapy. Clin. Cancer Res. (2001) 7(11):3314-3324.

104. SHAND N, WEBER F, MARIANI L et al.: A Phase 1-2 clinical trial of gene therapy for recurrent glioblastoma multiforme by tumor transduction with the herpes simplex thymidine kinase gene followed by ganciclovir. GLI328 European-Canadian Study Group. Hum. Gene Ther. (1999) 10(14):2325-2335.

105. HERMAN JR, ADLER HL, AGUILAR-CORDOVA E et al.: In situ gene therapy for adenocarcinoma of the prostate: a Phase I clinical trial. Hum. Gene Ther. (1999) 10(7):1239-1249.

106. SATOH T, TEH BS, TIMME TL et al.: Enhanced systemic T-cell activation after in situ gene therapy with radiotherapy in prostate cancer patients. Int. J. Radiat. Oncol. Biol. Phys. (2004) 59(2):562-571.

107. TEH BS, AYALA G, AGUILAR L et al.: Phase I-II trial evaluating combined intensity-modulated radiotherapy and in situ gene therapy with or without hormonal therapy in treatment of prostate cancer-interim report on PSA response and biopsy data. Int. J. Radiat. Oncol. Biol. Phys. (2004) 58(5):1520-1529.

108. STERMAN DH, TREAT J, LITZKY LA et al.: Adenovirus-mediated herpes simplex virus thymidine kinase/ganciclovir gene therapy in patients with localized malignancy: results of a Phase I clinical trial in malignant mesothelioma. Hum. Gene Ther. (1998) 9(7):1083-1092.

109. SHALEV M, KADMON D, TEH BS et al.: Suicide gene therapy toxicity after multiple and repeat injections in patients with localized prostate cancer. J. Urol. (2000) 163(6):1747-1750.

110. RAINOV NG: A Phase III clinical evaluation of herpes simplex virus Type 1 thymidine kinase and ganciclovir gene therapy as an adjuvant to surgical resection and radiation in adults with previously untreated glioblastoma multiforme. Hum. Gene Ther. (2000) 11(17):2389-2401.

111. JACOBS A, VOGES J, RESZKA R et al.: Positron-emission tomography of vector-mediated gene expression in gene therapy for gliomas. Lancet (2001) 358(9283):727-729.

•• PET study of the expression of transferred gene in GDEPT.

112. LOHR M, HOFFMEYER A, KROGER J et al.: Microencapsulated cell-mediated treatment of inoperable pancreatic carcinoma. Lancet (2001) 357(9268):1591-1592.

•• Use of allogenic cells as a vector in GDEPT.

113. PALMER DH, MAUTNER V, MIRZA D et al.: Virus-directed enzyme prodrug therapy: intratumoral administration of a replication-deficient adenovirus encoding nitroreductase to patients with resectable liver cancer. J. Clin. Oncol. (2004) 22(9):1546-1552.

114. PANDHA HS, MARTIN LA, RIGG A et al.: Genetic prodrug activation therapy for breast cancer: a Phase I clinical trial of erbB-2-directed suicide gene expression. J. Clin. Oncol. (1999) 17(7):2180-2189.

115. JAIN KK: Use of bacteria as anticancer agents. Expert Opin. Biol. Ther. (2001) 1(2):291-300.

116. SOGHOMONYAN SA, DOUBROVIN M, PIKE J et al.: Positron emission tomography (PET) imaging of tumor-localized Salmonella expressing HSV1-TK. Cancer Gene Ther. (2005) 12(1):101-108.

117. KING I, BERMUDES D, LIN S et al.: Tumor-targeted Salmonella expressing

cytosine deaminase as an anticancer agent. Hum. Gene Ther. (2002) 13(10):1225-1233.

118. BERMUDES D, ZHENG LM, KING IC: Live bacteria as anticancer agents and tumor-selective protein delivery vectors. Curr. Opin. Drug Disc. Dev. (2002) 5(2):194-199.

119. TJUVAJEV J, BLASBERG R, LUO X, ZHENG LM, KING I, BERMUDES D: Salmonella-based tumor-targeted cancer therapy: tumor amplified protein expression therapy (TAPET) for diagnostic imaging. J. Control. Release (2001) 74(1-3):313-315.

• Use of a bacterial delivery vector for diagnostic imaging.

120. BARNETT BG, CREWS CJ, DOUGLAS JT: Targeted adenoviral vectors. Biochim. Biophys. Acta (2002) 1575(1-3):1-14.

121. WEYEL D, SEDLACEK HH, MULLER R, BRUSSELBACH S: Secreted human beta-glucuronidase: a novel tool for gene-directed enzyme prodrug therapy. Gene Ther. (2000) 7(3):224-231.

122. HAMSTRA DA, LEE KC, TYCHEWICZ JM et al.: The use of 19F spectroscopy and diffusion-weighted MRI to evaluate differences in gene-dependent enzyme prodrug therapies. Mol. Ther. (2004) 10(5):916-928.

123. BROWN SB, BROWN EA, WALKER I: The present and future role of photodynamic therapy in cancer treatment. Lancet Oncol. (2004) 5(8):497-508.

124. SHARMAN WM, VAN LIER JE, ALLEN CM: Targeted photodynamic therapy via receptor mediated delivery systems. Adv. Drug Deliv. Rev. (2004) 56(1):53-76.

• Receptor-mediated delivery of photodynamic therapy.

125. JICHLINSKI P, LEISINGER HJ: Photodynamic therapy in superficial bladder cancer: past, present and future. Urol. Res. (2001) 29(6):396-405.

126. BARR H, KENDALL C, STONE N: Photodynamic therapy for esophageal cancer: a useful and realistic option. Technol. Cancer Res. Treat. (2003) 2(1):65-76.

127. BARBER P, BARR H, GEORGE J, KRASNER N, MORRIS AI, SUTEDJA TG: Photodynamic therapy in the treatment of lung and oesophageal cancers. Clin. Oncol. (R. Coll. Radiol.) (2002) 14(2):110-116.

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

iteit

Tw

ente

on

11/2

0/14

For

pers

onal

use

onl

y.

Page 15: Selective delivery of therapeutic agents for the diagnosis and treatment of cancer

Shukla & Krag

Expert Opin. Biol. Ther. (2006) 6(1) 53

128. O’NEAL DP, HIRSCH LR, HALAS NJ, PAYNE JD, WEST JL: Photo-thermal tumor ablation in mice using near infrared-absorbing nanoparticles. Cancer Lett. (2004) 209(2):171-176.

129. LOO C, LIN A, HIRSCH L et al.: Nanoshell-enabled photonics-based imaging and therapy of cancer. Technol. Cancer Res. Treat. (2004) 3(1):33-40.

• Use of nanoshell vectors for cancer imaging and treatment.

130. HAFELI UO: Magnetically modulated therapeutic systems. Int. J. Pharm. (2004) 277(1-2):19-24.

131. BABINCOVA M, LESZCZYNSKA D, SOURIVONG P, BABINEC P, LESZCZYNSKI J: Principles of magnetodynamic chemotherapy. Med. Hypotheses (2004) 62(3):375-377.

132. JORDAN A, SCHOLZ R, WUST P et al.: Effects of magnetic fluid hyperthermia (MFH) on C3H mammary carcinoma in vivo. Int. J. Hyperthermia (1997) 13(6):587-605.

133. JOHANNSEN M, THIESEN B, JORDAN A et al.: Magnetic fluid hyperthermia (MFH)reduces prostate cancer growth in the orthotopic Dunning R3327 rat model. Prostate (2005) 64(3):283-292.

134. MOROZ P, JONES SK, WINTER J, GRAY BN: Targeting liver tumors with hyperthermia: ferromagnetic embolization in a rabbit liver tumor model. J. Surg. Oncol. (2001) 78(1):22-29; discussion 30-21.

135. HILGER I, ANDRA W, HERGT R, HIERGEIST R, SCHUBERT H, KAISER WA: Electromagnetic heating of breast tumors in interventional radiology: in vitro and in vivo studies in human cadavers and mice. Radiology (2001) 218(2):570-575.

136. LUBBE AS, BERGEMANN C, RIESS H et al.: Clinical experiences with magnetic drug targeting: a Phase I study with 4’-epidoxorubicin in 14 patients with advanced solid tumors. Cancer Res. (1996) 56(20):4686-4693.

• Clinical study with magnetically targeted drug delivery in cancer patients.

137. WILSON MW, KERLAN RK JR, FIDELMAN NA et al.: Hepatocellular carcinoma: regional therapy with a magnetic targeted carrier bound to doxorubicin in a dual MR imaging/conventional angiography suite-initial experience with

four patients. Radiology (2004) 230(1):287-293.

•• Clinical study using a magnetic-targeted carrier bound to doxorubicin for selective delivery.

138. HALLAHAN DE, MAUCERI HJ, SEUNG LP et al.: Spatial and temporal control of gene therapy using ionizing radiation. Nat. Med. (1995) 1(8):786-791.

139. HALLAHAN DE, GENG L, CMELAK AJ et al.: Targeting drug delivery to radiation-induced neoantigens in tumor microvasculature. J. Control. Release (2001) 74(1-3):183-191.

140. HALLAHAN D, GENG L, QU S et al.: Integrin-mediated targeting of drug delivery to irradiated tumor blood vessels. Cancer Cell (2003) 3(1):63-74.

141. HALLAHAN DE, QU S, GENG L et al.: Radiation-mediated control of drug delivery. Am. J. Clin. Oncol. (2001) 24(5):473-480.

• A review on radiation-guided drug delivery technology.

142. UNGER EC, PORTER T, CULP W, LABELL R, MATSUNAGA T, ZUTSHI R: Therapeutic applications of lipid-coated microbubbles. Adv. Drug Deliv. Rev. (2004) 56(9):1291-1314.

143. HAUFF P, SEEMANN S, RESZKA R et al.: Evaluation of gas-filled microparticles and sonoporation as gene delivery system: feasibility study in rodent tumor models. Radiology (2005) 236(2):572-578.

144. YUH EL, SHULMAN SG, MEHTA SA et al.: Delivery of systemic chemotherapeutic agent to tumors by using focused ultrasound: study in a murine model. Radiology (2005) 234(2):431-437.

• Use of ultrasound-directed selective delivery of anticancer drugs to tumours.

145. HOSAIN F, SPENCER RP, COUTHON HM, STURTZ GL: Targeted delivery of antineoplastic agent to bone: biodistribution studies of technetium-99m-labeled gem-bisphosphonate conjugate of methotrexate. J. Nucl. Med. (1996) 37(1):105-107.

•• Bone-selective delivery of antineoplastic agents and imaging.

146. HIRABAYASHI H, FUJISAKI J: Bone-specific drug delivery systems: approaches via chemical modification of bone-seeking agents. Clin. Pharmacokinet. (2003) 42(15):1319-1330.

147. CHOURASIA MK, JAIN SK: Pharmaceutical approaches to colon

targeted drug delivery systems. J. Pharm. Pharm. Sci. (2003) 6(1):33-66.

148. ERION MD, VAN POELJE PD, MACKENNA DA et al.: Liver-targeted drug delivery using HepDirect prodrugs. J. Pharmacol. Exp. Ther. (2005) 312(2):554-560.

149. NERI D, BICKNELL R: Tumour vascular targeting. Nat. Rev. Cancer (2005) 5(6):436-446.

150. CHEN EX, SIU LL: Development of molecular targeted anticancer agents: successes, failures and future directions. Curr. Pharm. Des. (2005) 11(2):265-272.

151. LI KC, PANDIT SD, GUCCIONE S, BEDNARSKI MD: Molecular imaging applications in nanomedicine. Biomed. Microdevices (2004) 6(2):113-116.

152. NISHI Y: Enzyme/abzyme prodrug activation systems: potential use in clinical oncology. Curr. Pharm. Des. (2003) 9(26):2113-2130.

153. SHABAT D, LODE HN, PERTL U et al.: In vivo activity in a catalytic antibody-prodrug system: antibody catalyzed etoposide prodrug activation for selective chemotherapy. Proc. Natl. Acad. Sci. USA (2001) 98(13):7528-7533.

•• Use of Abzyme/catalytic antibody in enzyme prodrug therapy of cancer.

154. DESGRANGES C: [Monoclonal antibodies and therapeutics]. Pathol. Biol. (Paris) (2004) 52(6):351-364.

155. VATER A, KLUSSMANN S: Toward third-generation aptamers: Spiegelmers and their therapeutic prospects. Curr. Opin. Drug Disc. Dev. (2003) 6(2):253-261.

156. EULBERG D, KLUSSMANN S: Spiegelmers: biostable aptamers. Chembiochem (2003) 4(10):979-983.

157. REDDY JA, ALLAGADDA VM, LEAMON CP: Targeting therapeutic and imaging agents to folate receptor positive tumors. Curr. Pharm. Biotechnol. (2005) 6(2):131-150.

158. SHUKER SB, HAJDUK PJ, MEADOWS RP, FESIK SW: Discovering high-affinity ligands for proteins: SAR by NMR. Science (1996) 274(5292):1531-1534.

•• Small molecule ligand identification against protein targets.

159. ERLANSON DA, MCDOWELL RS, O’BRIEN T: Fragment-based drug discovery. J. Med. Chem. (2004) 47(14):3463-3482.

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

iteit

Tw

ente

on

11/2

0/14

For

pers

onal

use

onl

y.

Page 16: Selective delivery of therapeutic agents for the diagnosis and treatment of cancer

Selective delivery of therapeutic agents for the diagnosis and treatment of cancer

54 Expert Opin. Biol. Ther. (2006) 6(1)

160. ERLANSON DA, WELLS JA, BRAISTED AC: Tethering: fragment-based drug discovery. Annu. Rev. Biophys. Biomol. Struct. (2004) 33:199-223.

161. EDELSTEIN ML, ABEDI MR, WIXON J, EDELSTEIN RM: Gene therapy clinical trials worldwide 1989-2004-an overview. J. Gene Med. (2004) 6(6):597-602.

Website201. http://www.wiley.co.uk/wileychi/genmed/

clinical/Gene Therapy Clinical Trials Worldwide, website provided by The Journal of Gene Medicine, updated July 2005.

AffiliationGirja S Shukla†1 PhD & David N Krag1 MD†Author for correspondence1Vermont Comprehensive Cancer Center, Department of Surgery, University of Vermont College of Medicine, 89 Beaumont Avenue, Burlington, VT 05405, USATel: +1 802 656 9488; Fax: +1 802 656 5833;E-mail: [email protected]

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Uni

vers

iteit

Tw

ente

on

11/2

0/14

For

pers

onal

use

onl

y.