+ All Categories
Home > Documents > Liposomes and nanoparticles: nanosized vehicles for drug delivery in cancer

Liposomes and nanoparticles: nanosized vehicles for drug delivery in cancer

Date post: 22-Nov-2023
Category:
Upload: ucl
View: 0 times
Download: 0 times
Share this document with a friend
8
UNCORRECTED PROOF Liposomes and nanoparticles: nanosized vehicles for drug delivery in cancer Yogeshkumar Malam 1, 2 , Marilena Loizidou 2 and Alexander M. Seifalian 1, 2, 3 1 Centre for Nanotechnology, Biomaterials and Tissue Engineering, University College London, London, UK 2 UCL Division of Surgery & Interventional Science, University College London, London, UK 3 Royal Free Hampstead NHS Trust Hospital, London, UK Nanoscale drug delivery systems using liposomes and nanoparticles are emerging technologies for the rational delivery of chemotherapeutic drugs in the treatment of cancer. Their use offers improved pharmacokinetic prop- erties, controlled and sustained release of drugs and, more importantly, lower systemic toxicity. The commer- cial availability of liposomal Doxil W and albumin-nano- particle-based Abraxane W has focused attention on this innovative and exciting field. Recent advances in lipo- some technology offer better treatment of multidrug- resistant cancers and lower cardiotoxicity. Nanoparti- cles offer increased precision in chemotherapeutic tar- geting of prostate cancer and new avenues for the treatment of breast cancer. Here we review current knowledge on the two technologies and their potential applications to cancer treatment. Introduction The application of innovative nanotechnologies to medi- cine nanomedicine has the potential to significantly benefit clinical practice, offering solutions to many of the current limitations in diagnosis, treatment and manage- ment of human disease. The diverse branches of nanome- dicine include tissue regeneration [1], drug delivery [2] and imaging [3]. This review focuses on two nanotechnological drug delivery methods, liposomes and drug-conjugated nanoparticles. Liposomes are closed spherical vesicles consisting of a lipid bilayer that encapsulates an aqueous phase in which drugs can be stored. The liposome diameter varies from 400 nm to 2.5 mm. Nanoparticles (NPs), which are particles ranging in size from 1 to 100 nm, exhibit unique physical and chemical properties that can be exploited for drug delivery by conjugation with drugs. Both these emerging nanoscale drug delivery systems can be used to improve current treatment regimens (Box 1) Q1 . High drug toxicity is a barrier to treatment because side effects limit the drug dosage that can be administered. This is best exemplified by cytotoxic cancer drugs. Although very effective in vitro, in human clinical use the drugs act indiscriminately on both cancerous and healthy tissues. Side effects can be both serious and unpleasant and range from nausea and hair loss to neuropathies, neutropenia and kidney failure. Therefore, drug non-specificity limits efficacy [4]. Box 2 details recent drugs and diseases under investigation for the use of nanoscale drug delivery. This review outlines recent developments in the use of liposomes and NPs in the field of drug delivery for the treatment of cancer. An understanding of these new tech- nologies is needed for the advancement of chemotherapy with higher efficacy and lower toxicity. Advantages of nanoscale drug delivery systems The ideal nanoscale drug delivery system ensures that the conjugated or bound drugcarrier complex arrives and acts preferentially at the selected target. Targeting of the drugnanocarrier complex can be active, whereby the complex incorporates a ligand specific for the receptor or epitope of the target tissue (Table 1). In passive targeting, complexes diffuse and accumulate at sites with excessively leaky microvasculature, such as tumours and inflamed tissues, with normal endothelium being much less permeable. Subsequent extravasation of complexes takes place either via transcytosis, whereby macromolecules are internalized from the blood at points of invagination of the cell mem- brane, or paracellularly, via diffusion through the tight junctions of endothelial cells. Particularly in cancers, an imbalance in factors that regulate angiogenesis, such as overexpression of vascular endothelial growth factor (VEGF), results in both increased vascular permeability and chaotic tumour-vessel architecture. In combination, these effects cause enhanced permeation and retention (EPR) [5], resulting in high local drug concentrations. Key properties of any nanomaterial used in drug deliv- ery are its biocompatibility and biodegradability, so that the unloaded carrier is degraded or metabolized into non- toxic components and cleared through the circulation. Materials are cleared according to size. Small particles (030 nm) are rapidly cleared by renal excretion. Nanocar- riers >30 nm are cleared by the mononuclear phagocytic system (MPS), consisting of macrophages located in the liver (Kupffer cells) and the spleen [6], which act as pha- gocytotic scavengers. Clearance is also dependent on endo- thelial fenestral size [6]. Fenestrae are highly variable, so it is difficult to determine the efficacy and toxicity of nanomedicines in different individuals because age, sex and genetics influence their rate of clearance [4]. Whether nanocarriers are taken up by macrophages depends on Review 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 Corresponding author: Seifalian, A.M. ([email protected]). TIPS 737 1–8 0165-6147/$ see front matter ß 2009 Elsevier Ltd. All rights reserved. doi:10.1016/j.tips.2009.08.004 Available online xxxxxx 1
Transcript

Q1

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

21

22

23

24

25

26

27

28

29

30

31

32

33

34

35

36

37

38

39

40

41

42

43

44

45

46

47

48

49

50

51

52

53

54

55

56

57

58

59

60

61

62

63

64

65

66

67

68

69

70

71

72

73

74

75

76

77

TIPS 737 1–8

OF

Liposomes and nanoparticles:nanosized vehicles for drug deliveryin cancerYogeshkumar Malam1,2, Marilena Loizidou2 and Alexander M. Seifalian1,2,3

1 Centre for Nanotechnology, Biomaterials and Tissue Engineering, University College London, London, UK2 UCL Division of Surgery & Interventional Science, University College London, London, UK3 Royal Free Hampstead NHS Trust Hospital, London, UK

Review

C78

79

80

81

82

83

84

85

86

87

88

89

90

91

92

93

94

95

96

97

98

99

100

101

102

103

104

105

106

107

108

109

110

111

112

113

114

115

116

117

118

119

120

UN

CO

RR

ENanoscale drug delivery systems using liposomes andnanoparticles are emerging technologies for the rationaldelivery of chemotherapeutic drugs in the treatment ofcancer. Their use offers improved pharmacokinetic prop-erties, controlled and sustained release of drugs and,more importantly, lower systemic toxicity. The commer-cial availability of liposomal DoxilW and albumin-nano-particle-based AbraxaneW has focused attention on thisinnovative and exciting field. Recent advances in lipo-some technology offer better treatment of multidrug-resistant cancers and lower cardiotoxicity. Nanoparti-cles offer increased precision in chemotherapeutic tar-geting of prostate cancer and new avenues for thetreatment of breast cancer. Here we review currentknowledge on the two technologies and their potentialapplications to cancer treatment.

IntroductionThe application of innovative nanotechnologies to medi-cine – nanomedicine – has the potential to significantlybenefit clinical practice, offering solutions to many of thecurrent limitations in diagnosis, treatment and manage-ment of human disease. The diverse branches of nanome-dicine include tissue regeneration [1], drug delivery [2] andimaging [3]. This review focuses on two nanotechnologicaldrug delivery methods, liposomes and drug-conjugatednanoparticles.

Liposomes are closed spherical vesicles consisting of alipid bilayer that encapsulates an aqueous phase in whichdrugs can be stored. The liposome diameter varies from400 nm to 2.5 mm.Nanoparticles (NPs), which are particlesranging in size from 1 to 100 nm, exhibit unique physicaland chemical properties that can be exploited for drugdelivery by conjugation with drugs. Both these emergingnanoscale drug delivery systems can be used to improvecurrent treatment regimens (Box 1).

High drug toxicity is a barrier to treatment because sideeffects limit the drug dosage that can be administered. Thisis best exemplified by cytotoxic cancer drugs. Althoughvery effective in vitro, in human clinical use the drugs actindiscriminately on both cancerous and healthy tissues.Side effects can be both serious and unpleasant and rangefrom nausea and hair loss to neuropathies, neutropenia

121

122Corresponding author: Seifalian, A.M. ([email protected]).

0165-6147/$ – see front matter � 2009 Elsevier Ltd. All rights reserved. doi:10.1016/j.tips.2009.

TED

PR

Oand kidney failure. Therefore, drug non-specificity limitsefficacy [4]. Box 2 details recent drugs and diseases underinvestigation for the use of nanoscale drug delivery.

This review outlines recent developments in the use ofliposomes and NPs in the field of drug delivery for thetreatment of cancer. An understanding of these new tech-nologies is needed for the advancement of chemotherapywith higher efficacy and lower toxicity.

Advantages of nanoscale drug delivery systemsThe ideal nanoscale drug delivery system ensures that theconjugated or bound drug–carrier complex arrives and actspreferentially at the selected target. Targeting of the drug–

nanocarrier complex can be active, whereby the complexincorporates a ligand specific for the receptor or epitope ofthe target tissue (Table 1). In passive targeting, complexesdiffuse and accumulate at sites with excessively leakymicrovasculature, such as tumours and inflamed tissues,with normal endothelium being much less permeable.Subsequent extravasation of complexes takes place eithervia transcytosis, whereby macromolecules are internalizedfrom the blood at points of invagination of the cell mem-brane, or paracellularly, via diffusion through the tightjunctions of endothelial cells. Particularly in cancers, animbalance in factors that regulate angiogenesis, such asoverexpression of vascular endothelial growth factor(VEGF), results in both increased vascular permeabilityand chaotic tumour-vessel architecture. In combination,these effects cause enhanced permeation and retention(EPR) [5], resulting in high local drug concentrations.

Key properties of any nanomaterial used in drug deliv-ery are its biocompatibility and biodegradability, so thatthe unloaded carrier is degraded or metabolized into non-toxic components and cleared through the circulation.Materials are cleared according to size. Small particles(0–30 nm) are rapidly cleared by renal excretion. Nanocar-riers >30 nm are cleared by the mononuclear phagocyticsystem (MPS), consisting of macrophages located in theliver (Kupffer cells) and the spleen [6], which act as pha-gocytotic scavengers. Clearance is also dependent on endo-thelial fenestral size [6]. Fenestrae are highly variable, soit is difficult to determine the efficacy and toxicity ofnanomedicines in different individuals because age, sexand genetics influence their rate of clearance [4]. Whethernanocarriers are taken up by macrophages depends on

08.004 Available online xxxxxx 1

123

124

125

126

127

128

129

130

131

132

133

134

135

136

137

138

139

140

141

142

143

144

145

146

147

148

149

150

151

152

153

154

155

156

157

158

159

160

161

162

163

164

165

166

167

168

169

170

171

172

173

174

175

176

177

178

179

180

181

182

183

184

185

186

187

188

189

190

191

192

193

194

195

196

197

198

199

200

201

202

203

204

205

206

207

208

209

210

211

212

213

214

215

216

217

218

219

220

221

222

Box 1. Goals of a nanoscale drug delivery system

(i) Targeting, to increase the drug concentration at desired sites of

action and reduce systemic levels of the drug and its toxic

sequelae in healthy tissues.

(ii) Improved solubility, to facilitate parenteral drug administration.

(iii) Constant rate of drug delivery, resulting in zero-order release

kinetics to maintain a constant therapeutic dose at the site of

action [9].

(iv) Reduced clearance, to increase the drug half-life.

(v) Increased drug stability, to reduce degradation and maximize

drug action.

(vi) Drug delivery across the blood–brain barrier (BBB) [58] and

blood–cochlear barrier [59].

Review Trends in Pharmacological Sciences Vol.xxx No.x

TIPS 737 1–8

RE

C

opsonization by the innate immune system [7]. Opsonins,molecules that bind to foreign materials and enhancephagocytosis, include IgG and IgA antibodies, the comp-lement cascade system and mannose-binding lectin [8].Therefore, the surface properties of nanocarriers can sig-nificantly affect the rate of clearance by the MPS. A usefulmethod for evading opsonization of large narrow carrierswas developed in Rutgers University in the 1960s [9]: in aprocess called PEGylation, a polymer, poly(ethylene glycol)(PEG; [CH2CH2O]n), is conjugated to the drug carrier.

Overall, use of ligand–drug–nanocarrier complexesimproves the drug therapeutic index according toEq. (1). The high selectivity and specificity of the complexincrease the amount of drug delivered to the target tissueand decrease the amount at unwanted sites. Therefore,less systemic drug needs to be administered to ensure asufficient concentration at the site of action and the mini-mum efficacious dose is also lower. In addition, becauseless drug is present at unwanted sites, the maximum non-toxic is higher. The overall effect is a drastic decrease intoxicity and adverse side effects [10].

Therapeutic index ¼Maximum non� toxic dose

Minimum effective dose: (1)

UN

CO

RBox 2. Potential therapeutic opportunities for nanoscale drug de

Nanocarrier Drug Disease

SLNs [60] Insulin Diabetes mellitus

Liposomes [61] Vasoactive

intestinal

peptide (VIP)

Hypertension

Liposomes

(Ambisome1) [62]

Amphotericin B Fungal infections

Gold nanoparticles [63] Ciprofloxcain Bacterial infections

such as urinary tract

infections, cystitis,

sinusitis and respirator

tract infections

PLGA nanoparticles [64] Rifampicin Tuberculosis

PLGA nanoparticles [65] Benzocaine Pain relief

SLNs [66] Clozapine Schizophrenia

2

TED

PR

OO

F

Nanoscale drug delivery systemsLiposomes

The liposome bilayer can be composed of either synthetic ornatural phospholipids. The predominant physical andchemical properties of a liposome are based on the netproperties of the constituent phospholipids [11], includingpermeability, charge density and steric hindrance. Thelipid bilayer closes in on itself due to interactions betweenwater molecules and the hydrophobic phosphate groups ofthe phospholipids. This process of liposome formation isspontaneous because the amphiphilic phospholipids self-associate into bilayers. Drug loading into liposomes can beachieved through (i) liposome formation in an aqueoussolution saturated with soluble drug; (ii) the use of organicsolvents and solvent exchange mechanisms; (iii) the use oflipophilic drugs; and (iv) pH gradient methods [12](Figure 1).

Liposomes generally reach their site of action by extra-vasation into the interstitial space from the bloodstream.Liposomes can target specific tissues through both activeand passive targeting strategies (Figure 2). This is becauseliposomes can easily be manipulated by adding additionalmolecules to the outer surface of the lipid bilayer. Becauseliposomes are of the order of 400 nm in size, they arerapidly cleared by the MPS system. Reducing opsonizationof liposomes by PEGylation therefore reduce clearance bythe MPS, increasing the circulation half-life. Opsonizationpresents such a problem to the development of therapeuti-cally useful liposomes that nearly all research reported inthe literature involves PEG-coated or PEGylated lipo-somes.

Liposomal formulations of anticancer drugs havealready been approved for human use. Doxil1 is a liposo-mal formulation of the anthracycline drug doxorubicinused to treat cancer in AIDS-related Kaposi sarcomaand multiple myeloma [13]. Its advantages over free dox-orubicin are greater efficacy and lower cardiotoxicity.These advantages are attributed to passive targeting of

223

224

225

226

227

228

229

230

231

232

233

234

235

236

237

238

239

240

241

242

243

244

livery in diseases other than cancer

Advantages

Pulmonary administration possible; an inhaler

or nebulizer replaces a daily regimen of subcutaneous

injections, increasing patient satisfaction and compliance

Potential new treatment for hypertension using VIP, which

is limited by rapid degradation in blood by first-pass hepatic

circulation

Reduced renal toxicity and greater efficacy in treating

fungal infections; also used to treat other parasitic infections

y

Sustained release over a number of hours and greater local

concentrations of the free drug at sites of pathology because

of the permeation and retention effect

Sustained release over a period of days, increasing patient

compliance because medication can be taken weekly instead

of daily over a period of 6 months

Parental administration possible; only a single dose is required

for a prolonged effect

Higher clozapine concentrations across the blood–brain

barrier compared to clozapine solution

C

OO

F

245

246

247

248

249

250

251

252

253

254

255

256

257

258

259

260

261

262

263

264

265

266

267

268

269

270

271

272

273

274

275

276

277

278

279

280

281

282

283

284

285

286

287

288

289

290

291

292

293

294

295

296

297

298

299

300

301

302

303

304

305

306

307

308

309

310

311

312

313

314

315

316

317

318

319

320

321

322

323

324

325

326

327

328

329

330

331

332

333

334

335

336

337

338

339

340

341

342

343

344

345

346

Table 1. Typical examples of active targeting with drug delivery systemsa.

Ligand Receptor/target Study findings

Anti-CD74 antibody, LL1 [49] CD74 receptor Ligand covalently attached to liposomes; selective for malignant B lymphoma cells

TfR-targeting peptide

HAIYPRH [50]

TfR Conjugation to the TfR-binding peptide significantly improves the anti-cancer

potency and selectivity of the anti-cancer drug artemisinin

Folate [51] Folate receptor (FR) FR is overexpressed on cancer cells

Folate has been conjugated on liposomes loaded with doxorubicin for targeting of

cancer an on NPs for targeted paclitaxel delivery

mBAFF [52] BAFF receptor BAFF is the usual endogenous ligand for the BAFF receptor; mBAFF is a soluble BAFF

mutant in which amino acids 217–224 are replaced by two glycine residues that can

bind to BAFF receptors

PEGylated liposomes developed with mBAFF as a targeting ligand target certain

B lymphoma cells in vitro

Hyaluronic acid [53] Hyaluronan

receptors (HR)

HT-29 cancer cells overexpress HR

Hyaluronic acid incorporated in chitosan NPs loaded with the anti-cancer drug

5-flurouracil exhibited higher cytotoxicity in vitro

Galactose [54] ASGP receptors ASGP receptors are overexpressed on hepatoma cells

Dextran-based polymeric micelles were used to target liver cancer in vivo with

superior resultsaAGIP, amyloid growth inhibitory peptide; ASGP, asialo glycoprotein; mBAFF, mutant B cell activating factor belonging to the TNF family; SAP, sweet arrow peptide; TfR,

transferrin receptor.

Review Trends in Pharmacological Sciences Vol.xxx No.x

TIPS 737 1–8

RE

tumours, due to leaky tumour vasculature [14] and theEPR effect, and to lower concentrations of free doxorubicinat healthy tissue sites. There is evidence that liposomalDoxil1 is metabolized by leukaemia cells via a differentmechanism than that for free doxorubicin, which mightexplain the improved efficacy and lower toxicity. Further-more, Doxil1 is under clinical trial for the treatment ofbreast cancer.

One of the most interesting developments in this field isthe potential of solid lipid NPs to combat the increasingproblem of multidrug resistance (MDR) acquired by can-cers, which drastically reduces chemotherapeutic efficacy.Proposed mechanisms underlying MDR at the cellularlevel include: (i) increased metabolism of drugs due toincreased enzyme expression, especially of glutathioneS-transferase; (ii) drug transporters and efflux proteins[15]; and (iii) point mutations in proteins that are thera-peutic or drug targets. Ogawara et al. recently investigatedthe effect of PEG liposomal doxorubicin (Doxil1) in a malemouse tumour model inoculated with either colon cancer(C26) cells or their doxorubicin-resistant (MDR) subclone,

UN

CO

R

Figure 1. Diagram of a bilaminar liposome. The hydrophobic region traps drugs in

functionalized with ligands for active targeting or PEGylated. Liposomes can vary in the

multilamellar vesicles, (ii) large unilamellar vesicles and (iii) small unilamellar vesicles.

TED

PR

which overexpresses P-gp efflux pumps [16]. The resultsshowed that PEG liposomal doxorubicin had anti-tumoureffects on both doxorubicin-resistant and non-doxorubicin-resistant C26 cells. With increasing incidence of resistanceto chemotherapy, the use of liposomes offers effectivetreatment without the need for the costly discovery ofnew chemotherapeutic drugs because current drugs canbe reformulated.

To date, no specific in vivo study has compared theefficacy of liposomes to that of other nanoparticle deliverysystems; therefore, we cannot comment on the relativeefficacy of liposomes.

Liposomes are firmly established with the success ofDoxil1 and liposomal formulations of other anticancerdrugs are now being intensively explored to improve che-motherapy outcomes and reduce toxicity.

Solid lipid NPs

Solid lipid NPs (SLNs), also referred to as lipospheresor solid lipid nanospheres, are solid lipids at humanphysiological temperature (37 8C) and have a diameter

347

348

349

350

351

352

353

354

355

356

357

358

359

360

361

362

363

364

365

366

the central core when the liposomes are prepared. The outer surface can be

number of lipid bilayers they possess and can be classified into three categories: (i)

3

UN

CO

RR

EC

TED

PR

OO

F

367

368

369

370

371

372

373

374

375

376

377

378

379

380

381

382

383

384

385

386

387

388

389

390

391

392

393

394

395

396

397

398

399

400

401

402

403

404

405

406

407

408

409

410

411

412

413

414

415

416

417

418

419

420

421

422

423

424

425

426

427

428

429

430

431

432

433

434

435

436

437

438

439

440

441

442

443

444

445

446

447

448

449

450

451

452

453

454

455

456

457

458

459

460

461

462

463

464

465

466

467

468

469

470

471

472

473

474

475

476

477

478

479

480

481

482

483

484

485

486

487

488

Figure 2. Active and passive targeting of cells for drug targeting using liposomes. At sites of pathology where the endothelium layer is inflamed, mediators such as

bradykinin, vascular endothelium growth factor and prostaglandins increase the endothelial permeability. Underlying pathology includes cancer, rheumatoid arthritis and

infection. Liposomes extravasate through the gaps between cells and enter the interstitial fluid. Active targeting is achieved by conjugating ligands to the liposome that

bind to a specific target cell receptor, leading to internalization or release of the drug. Passive targeting can be mediated by internalization or local high-concentration

release of the drug. Adapted from Ref. [32]Q3 .

Figure 3. Benefits of SLNs in doxorubicin delivery. The cytotoxicity of free

doxorubicin, doxorubicin-loaded SLNs and unloaded SLNs at different

concentrations towards HT-29 colorectal cancer cells after 72-h exposure is

shown. Doxorubicin-loaded SLNs showed the highest toxicity, offering more

potent treatment than conventional free doxorubicin. Unloaded SLNs did not

induce any significant toxicity, which confirms that they are a safe carrier in vitro

[19].

Review Trends in Pharmacological Sciences Vol.xxx No.x

TIPS 737 1–8

4

of 50–1000 nm. They can be formed from a range of lipids,including mono-, di- and triglycerides, fatty acids, waxesand combinations thereof. SLNs are produced by replacingthe liquid lipid (oil) of an oil-in-water emulsion by a solidlipid and many commercially viable methods are availablefor large-scale production. SLNs are biodegradable andbiocompatible and can be used in humans because of theirlow toxicity [17]. SLNs must be stabilized by surfactants toform administrable emulsions [18].

SLNs form a strongly lipophilic matrix into which drugscan be loaded for subsequent release. The principal factorsaffecting drug loading into the SLN matrix are: (i) thesolubility of the drug in lipid (the drug must be lipophilic);(ii) the chemical and physical properties of the lipid ormixture of lipids; (iii) the crystalline characteristics of thelipid(s) at biological temperature; and (iv) the polymorphicform of the lipid(s) used. Use of a heterogeneous lipidmixture promotes an imperfect crystalline structure withlarger gaps for superior drug loading.

SLNs have been investigated for the delivery of variousanticancer drugs, with promising results in preclinicalmouse trials specifically showing that SLNs might helpto overcome MDR in cancers [17]. Serpe et al., using coloncancer cells in vitro, demonstrated the benefits of SLNs inthe delivery of doxorubicin (Figure 3), cholesteryl butyrateand paclitaxel [19].

C

OF

489

490

491

492

493

494

495

496

497

498

499

500

501

502

503

504

505

506

507

508

509

510

511

512

513

514

515

516

517

518

519

520

521

522

523

524

525

526

527

528

529

530

531

532

533

534

535

536

537

538

539

540

541

542

543

544

545

546

547

548

549

550

551

552

553

554

555

556

557

558

559

560

561

562

563

564

565

566

567

568

569

570

571

572

573

574

575

576

577

578

579

580

581

582

583

584

585

586

587

588

589

590

591

592

593

594

595

596

597

598

599

600

601

602

603

604

605

606

607

608

609

610

Table 2. Representative examples of studies using drug-carrying nanoparticlesa

NP polymer Drug Study findings

PLGA [55] Doxorubicin A single intravenous injection of doxorubicin conjugated to PLGA NP exhibited tumour

suppression comparable to that by daily injection of free doxorubicin over 12 days; thus,

the NP formulation was much more potent and longer-lasting than conventional free

doxorubicin

PLGA [51] Dexamethasone A single administration produced at least 14 days of sustained drug release; clinical

application in suppressing glial cell proliferation on implanted electrodes for

neurophysiological investigations into neural activity

PLA [56] Thyrotropin-

releasing

hormone

Intranasal delivery through olfactory neurons to reach the brain; tested for an anticonvulsant

in an animal seizure model; clinical application of peptide delivery to the brain without

crossing the blood–brain barrier

PLA–TPGS/MMT NP [57] Docetaxel Much greater cytotoxic potency to cancer cells than Taxotere1 (current clinical form of docetaxel)aNP, nanoparticle; PLA–TPGS/MMT NP, poly(lactide)–D-a-tocopheryl poly(ethylene glycol) 1000 succinate copolymer incorporated in montmorillonite medical clay.

Review Trends in Pharmacological Sciences Vol.xxx No.x

TIPS 737 1–8

UN

CO

RR

EIn an exciting development, mitoxantrone, a topoisome-

rase inhibitor that blocks DNA replication, was loaded intoSLNs and used in vivo as a local injection to treat breastcancer and lymph node metastases in mice [20]. Theresults revealed a nearly threefold reduction in lymphnode size compared to free mitoxantrone, which is a sig-nificant improvement on the existing treatment.

SLNs offer an alternative platform for drug delivery incancer. However, more in vivo studies are required beforethey can be translated to human treatment.

Polymer-based NPs

PolymericNPs have been extensively investigated as drugnanocarriers. As a class ofmolecule, their designs are verysimilar, with a polymeric backbone – usually formed froma biodegradable monomer based on a simple organic mol-ecule that is biocompatible – and functional moieties foractive targeting intercalated into the structure [21]. Drugloading is achieved either by (i) entrapment of an aqueousdrugphaseusing thepolymer to formnanoscale structuressuch as cages and capsules [21,22] or (ii) chemical linkingof the drugmolecules to the polymer backbone bymeans ofa simple ester or amide bond that can be hydrolyzed invivo. More complex polymeric NPs use polar groups tocreate hydrophobic and hydrophilic regions enable allowthe drug to adsorb onto the NP and facilitate delivery tothe target site.

The most widely researched synthetic polymers includepolylactide (PLA) [23], poly(D,L-lactide–co-glycolide)(PLGA) [24] and PEG [25]. All three polymers are hydro-lyzed in vivo and are biodegradable. Other polymers basedon biological polysaccharides have been extensively inves-tigated, including chitosan, cyclodextrin and dextrans [26].Different polymers can be combined to form co-polymers.PLA-block-PEG co-polymers harness the properties of bothpolymers, especially the anti-opsonization of PEG [24].Ligands can be attached to the NP to facilitate activetargeting. Ligands can be intercalated into the structureeither by direct covalent linkage to the polymeric backboneor through the use of biologically inert spacer groups [27].

Ligands for active targeting of cancer are used to exploitany specific antigens expressed by cancer cells. RNA A10aptamers specific for the prostrate-specificmembrane anti-gen have been successfully conjugated onto PLA-block-PEG co-polymers, which exhibited increased drug deliveryto prostate tumour cells compared to non-targeting NPs

TED

PR

O[27]. This is a promising development and might offerbetter non-surgical treatment for prostate cancer patients.

Current paclitaxel formulations (Taxol1), a drug forbreast cancer chemotherapy, use the organic solvent Cre-mophor EL1, which can elicit severe hypersensitivity reac-tions. PEGylated PLGA copolymer NPs showed anencapsulation efficiency of 70% for paclitaxel and induceda similar level of apoptotic cell death as that observed forTaxol1 when tested on HeLa cancer cells [28]. Signifi-cantly, the PEGylated PLGA copolymer showed no toxicityand therefore an effective formulation of paclitaxel can beproduced without the adverse effects associated with Cre-mophor EL1 [28]. This offers an alternative treatment tothose who are sensitive to Cremophor EL1 without com-promising on chemotherapeutic potency.

Cisplatin, another anticancer agent, has been loadedinto copolymer PLGA–methoxy-PEG (PLGA–mPEG) NPs[29]. In vitro testing revealed that cisplatin-loaded PLGA–

mPEG NPs passively targeted LNCaP prostate cancercells. Cisplatin-loaded NPs evoked less cytotoxicity thatfree cisplatin solution, but their passive targeting reducedsystemic toxicity. Fluorescence microscopy revealed thatcisplatin-loaded NP uptake occurred via internalization.An in vivomousemodel revealed that cisplatin blood levelswere prolonged and sustained at therapeutic concen-trations after intravenous administration. Table 2 listssome other salient studies in this field.

Polymeric NPs are still in the preclinical phase of de-velopment but have potential for the targeted delivery ofanticancer drugs owing to ease with which ligands can beattached.

Gold NPs

Gold NPs consist of a core gold atom that can be functio-nalized by addition of a monolayer of moieties containing athiol (SH) group [30]. Examples of these moieties includeligands for active targeting of the gold NP, such as maskedphosphonioalkyl selenoates [31], peptides and glyconano-particles. Gold NPs can be synthesized using NaBH4 toreduce AuCl4

� salts in the presence of thiol-containingmoieties that subsequently form a monolayer around thecore gold atom, depending on the stoichiometric gold/thiolratio (Figure 4) [32]. Synthesized NPs have a diameter of1–150 nm. Further NP modification can be carried outusing a place exchange reaction, in which thiol-containingmoieties are swapped. In this way, a single gold NP core

5

F

611

612

613

614

615

616

617

618

619

620

621

622

623

624

625

626

627

628

629

630

631

632

633

634

635

636

637

638

639

640

641

642

643

644

645

646

647

648

649

650

651

652

653

654

655

656

657

658

659

660

661

662

663

664

665

666

667

668

669

670

671

672

673

674

675

676

677

678

679

680

681

682

683

684

685

686

687

688

689

690

691

692

693

694

695

696

697

698

699

700

701

702

703

704

705

706

707

708

709

710

711

712

713

714

715

716

717

718

719

720

721

722

723

724

725

726

727

728

729

730

731

732

Figure 4. Synthesis of gold nanoparticles. In step 1, the Schiffrin reaction, AuCl4� is reduced by NaBH4 in the presence of functional moieties with thiol groups. In step 2, the

Murray reaction, different functional moieties with thiol groups (represented by different colours) can be swapped in a place-exchange reaction [32]. Step 3 involves further

addition of a different thiol ligand.

Review Trends in Pharmacological Sciences Vol.xxx No.x

TIPS 737 1–8

UN

CO

RR

EC

can be functionalized with many different groups for tar-geting, stability, evasion of host defences and drug delivery[32].

Studies have confirmed that gold NPs are non-toxic atthe cellular level in a number of human cell lines [33].Studies in mice using gold NPs as an imaging agentrevealed no evidence of toxicity over 30 days [34]. A pio-neering study demonstrated that PEGylated gold NPs (10–

30 nm) are unable to cross the human placenta within 6 h,which could be used to restrict drug delivery to just themother while preventing teratogenic effects on the foetus[35].

Drug delivery using gold NPs is still in its infancy,although much more progress has been made in DNAdelivery for gene therapy [36] and in imaging [37]. GoldNPs can be synthesized and functionalized with anticancerdrugs such as paclitaxel and 6-mercaptopurine (6-MP) [38].Gold NPs co-administered with paclitaxel show enhancedanti-proliferation effects on tumours. It is thought thatgold NPs disrupt cell adhesion [39]. The anti-leukaemiadrug 6-MP bound to gold NPs exhibits greater in vitrotoxicity against leukaemia than free 6-MP, even thoughgold NPs had no anti-leukaemia activity in control studies.

The most novel development for gold NPs is the use ofintracellular glutathione as a trigger for drug release [40].The higher glutathione levels found in cancerous and pre-cancerous cells could thus be exploited in targeting intra-cellular release of chemotherapy drugs [41].

Although drug delivery using gold NPs is still evolving,there is potential for developing multifunctional particlesfor imaging, drug and gene delivery systems for applicationin cancer.

Albumin NPs

Albumin, a plasma protein with a molecular weight of 66kDa, has been extensively investigated as a drug carrier,with promising results. It is soluble in both water andethanol, two viable solvents for intravenous adminis-tration. Because albumin is found in the circulatingplasma of the human body at concentrations of 50 g/L ofserum, it is non-toxic and well tolerated by the immunesystem. Albumin has favourable pharmacokinetics owingto its long half-life in circulating plasma, which makes itparticularly attractive as a drug carrier for passive target-ing [42]. Albumin can be derived from human plasma andblood products. Alternatively, recombinant human serumalbumin can be produced in genetically engineered yeastcells [42]. Albumin NPs are prepared by desolvation orcoacervation.

6

TED

PR

OOAbraxane1, also known as nab-paclitaxel, was the first

drug based on an albumin NP approved for human use bythe US Food and Drug Administration. The chemotherapydrug paclitaxel is bound to 130-nm human albumin NPs.Abraxane1 has advantages over free paclitaxel in terms ofits longer circulation half-life and lack of the hypersensi-tivity-inducing Cremophor EL1 solvent [43]. Clinical trialshave confirmed the efficacy of Abraxane1 in the treatmentof metastatic breast cancer, for which it is routinely used[44]. In addition, Abraxane1 is currently being investi-gated with other taxanes in the treatment of hormonerefractory prostate cancer [44]. Albumin is transportedacross the endothelium into the extravascular space bytranscytosis via caveolae, initiated by the albumin receptorgp60 [45]. Tumour tissues have a high metabolic demandand actively transport plasma proteins into their cells foranabolic processes. It has been proposed that this mech-anism would explain why Abraxane1 targets and prefer-entially accumulates in cancer tissues in vivo [46] via theexcessive vascular network associated with cancers [44].There is also speculation that Abraxane1 is transportedinto tumour cells by secreted protein acidic rich in cysteine(SPARC) or osteonectin [44].

Albumin–PEG–PLA NPs cross the blood–brain barrier[47] and conjugation of apolipoproteins can facilitate trans-cytosis [48]. These findings open new avenues for the use ofalbumin NPs as a carrier for drug delivery to the brain notonly for cancer treatment, but also for a wide range ofcentral nervous system diseases.

Conclusions and future directionLiposomes and NPs are promising candidates for the de-velopment of drug delivery systems. Early experimentalevidence, both clinically and preclinically, shows greatpotential for the widespread adoption of liposomes andNPs in cancer treatment. Their attractive properties in-clude biocompatibility, low toxicity, lower clearance rates,the ability to target specific tissues and controlled releaseof drugs. They offer numerous advantages over conven-tional chemotherapy using free drug treatment, as evi-denced by the approval of Abraxane1 and Doxil1. Both ofthese nanomaterial-based formulations of existing drugsoffer better pharmacokinetic properties and lower systemictoxicity of the chemotherapeutic drugs that they deliver.

However, the full potential of these emerging technol-ogies has not yet been fully realized. The toxicology ofnanomaterials in humans still needs to be fully studiedand evaluated. Studies so far have been small and limitedto short-term exposure; few have looked at the wider

C

733

734

735

736

737

738

739

740

741

742

743

744

745

746

747

748

749

750

751

752

753

754

755

756

757

758

759

760

761

762

763

764

765

766

767

768

769

770

771

772

773

774

775

776

777

778

779

780

781

782

783

784

785

786

787

788

789

790

791

792

793

794

795

796

797

798

799

800

801

802

803

804

805

806

807

808

809

810

811

812

813

814

815

816

817

818

819

820

821

822

823

824

825

826

827

828

829

830

831

832

833

834

835

836

837

838

839

840

841

842

843

844

845

846

847

848

849

850

851

852

853

Review Trends in Pharmacological Sciences Vol.xxx No.x

TIPS 737 1–8

UN

CO

RR

E

impact. Investigation into so-called nanotoxicity shouldfocus on long-term exposure in humans, animals and theenvironment. Further in vivo studies are needed to deter-mine the efficacy of these new drug formulations, culmi-nating in phase I trials. The reproducibility of batches ofdrug formulations such as liposomes and NPs also needs tobe refined.

Liposomes and NPs are just beginning to make animpact in chemotherapy owing to the dual drive to reducethe toxicity and side effects of existing treatments andincrease efficacy by selective targeting of tumours.

AcknowledgementWe would like to thank the EPSRC for financial support for thedevelopment of nanoparticles and nanomaterials for drug delivery andthe NHIR for a (Neat) grant.

References1 Zhang, L. and Webster, T.J. (2009) Nanotechnology and

nanomaterials: promises for improved tissue regeneration. NanoToday 4, 66–80

2 Yokoyama, M. (2005) Drug targeting with nano-sized carrier systems.J. Artif. Organs 8, 77–84

3 Jamieson, T. et al. (2007) Biological applications of quantum dots.Biomaterials 28, 4717–4732

4 Igarashi, E. (2008) Factors affecting toxicity and efficacy of polymericnanomedicines. Toxicol. Appl. Pharmacol. 229, 121–134

5 Yaghini, E. et al. (2009) Quantum dots and their potential biomedicalapplications in photosensitization for photodynamic therapy.Nanomedicine 4, 353–363

6 Gaumet, M. et al. (2008) Nanoparticles for drug delivery: the need forprecision in reporting particle size parameters. Eur. J. Pharm.Biopharm. 69, 1–9

7 Owens, D.E., III and Peppas, N.A. (2006) Opsonization, biodistribution,and pharmacokinetics of polymeric nanoparticles. Int. J. Pharm. 307,93–102

8 Nagayama, S. et al. (2007) Time-dependent changes in opsonin amountassociated on nanoparticles alter their hepatic uptake characteristics.Int. J. Pharm. 342, 215–221

9 Hoffman, A.S. (2008) The origins and evolution of ‘‘controlled’’ drugdelivery systems. J. Control. Release 132, 153–163

10 Vega-Villa, K.R. et al. (2008) Clinical toxicities of nanocarrier systems.Adv. Drug Deliv. Rev. 60, 929–938

11 Bawarski, W.E. et al. (2008) Emerging nanopharmaceuticals.Nanomedicine 4, 273–282

12 Qiu, L. et al. (2008) Preparation and in vitro evaluation of liposomalchloroquine diphosphate loaded by a transmembrane pH-gradientmethod. Int. J. Pharm. 361, 56–63

13 Ning, Y.M. et al. (2007) Liposomal doxorubicin in combination withbortezomib for relapsed or refractory multiple myeloma. Oncology(Williston Park) 21, 1503–1508

14 Ogawara, K.i. et al. (2008) Determinants for in vivo anti-tumor effectsof PEG liposomal doxorubicin: importance of vascular permeabilitywithin tumors. Int. J. Pharm. 359, 234–240

15 Higgins, C.F. (2007) Multiple molecular mechanisms for multidrugresistance transporters. Nature 446, 749–757

16 Ogawara, K.i. et al. (2009) In vivo anti-tumor effect of PEG liposomaldoxorubicin (DOX) inDOX-resistant tumor-bearingmice: involvement ofcytotoxic effect onvascularendothelial cells.J.Control.Release133, 4–10

17 Wong, H.L. et al. (2007) Chemotherapy with anticancer drugsencapsulated in solid lipid nanoparticles. Adv. Drug Deliv. Rev. 59,491–504

18 Pardeike, J. et al. (2009) Lipid nanoparticles (SLN, NLC) in cosmeticand pharmaceutical dermal products. Int. J. Pharm. 366, 170–184

19 Serpe, L. et al. (2004) Cytotoxicity of anticancer drugs incorporated insolid lipid nanoparticles on HT-29 colorectal cancer cell line. Eur. J.Pharm. Biopharm. 58, 673–680

20 Lu, B. et al. (2006) Solid lipid nanoparticles of mitoxantrone for localinjection against breast cancer and its lymph node metastases. Eur. J.Pharm. Sci. 28, 86–95

TED

PR

OO

F

21 Bajpai, A.K. et al. (2008) Responsive polymers in controlled drugdelivery. Prog. Polym. Sci. 33, 1088–1118

22 Deng, X. et al. (2007) Translocation and fate of multi-walled carbonnanotubes in vivo. Carbon 45, 1419–1424

23 Hu, K. et al. (2009) Lactoferrin-conjugated PEG-PLA nanoparticleswith improved brain delivery: in vitro and in vivo evaluations. J.Control. Release 134, 55–61

24 Cheng, J. et al. (2007) Formulation of functionalized PLGA–PEGnanoparticles for in vivo targeted drug delivery. Biomaterials 28,869–876

25 Chan, J.M. et al. (2009) PLGA–lecithin–PEG core–shell nanoparticlesfor controlled drug delivery. Biomaterials 30, 1627–1634

26 Liu, Z. et al. (2008) Polysaccharides-based nanoparticles as drugdelivery systems. Adv. Drug Deliv. Rev. 60, 1650–1662

27 Byrne, J.D. et al. (2008) Active targeting schemes for nanoparticlesystems in cancer therapeutics. Adv. Drug Deliv. Rev. 60, 1615–1626

28 Danhier, F. et al. (2009) Paclitaxel-loaded PEGylated PLGA-basednanoparticles: in vitro and in vivo evaluation. J. Control. Release133, 11–17

29 Gryparis, E.C. et al. (2007) Anticancer activity of cisplatin-loadedPLGA–mPEG nanoparticles on LNCaP prostate cancer cells. Eur. J.Pharm. Biopharm. 67, 1–8

30 Xu, Z.P. et al. (2006) Inorganic nanoparticles as carriers for efficientcellular delivery. Chem. Eng. Sci. 61, 1027–1040

31 Ju-Nam, Y. et al. (2007) The synthesis and characterisation of maskedphosphonioalkyl selenoates: potential ligands for the production offunctionalised gold nanoparticles. J. Organomet. Chem. 692, 5065–5070

32 Ghosh, P. et al. (2008) Gold nanoparticles in delivery applications. Adv.Drug Deliv. Rev. 60, 1307–1315

33 Connor, E.E. et al. (2005) Gold nanoparticles are taken up by humancells but do not cause acute cytotoxicity. Small 1, 325

34 Hainfeld, J.F. et al. (2006) Gold nanoparticles: a new X-ray contrastagent. Br. J. Radiol. 79, 248–253

35 Myllynen, P.K. et al. (2008) Kinetics of gold nanoparticles in the humanplacenta. Reprod. Toxicol. 26, 130–137

36 Lee, S.H. et al. (2008) Amine-functionalized gold nanoparticles as non-cytotoxic and efficient intracellular siRNA delivery carriers. Int. J.Pharm. 364, 94–101

37 Huang, X. et al. (2007) Gold nanoparticles: interesting opticalproperties and recent applications in cancer diagnostics andtherapy. Nanomedicine 2, 681

38 Podsiadlo, P. et al. (2007) Gold nanoparticles enhance the anti-leukemia action of a 6-mercaptopurine chemotherapeutic agent.Langmuir 24, 568–574

39 Wei, X.L. et al. (2007) Disruption of HepG2 cell adhesion by goldnanoparticle and Paclitaxel disclosed by in situ QCM measurement.Colloids Surf. B Biointerfaces 59, 100–104

40 Hong, R. et al. (2006) Glutathione-mediated delivery and release usingmonolayer protected nanoparticle carriers. J. Am. Chem. Soc. 128,1078–1079

41 Estrela, J.M. et al. (2006) Glutathione in cancer biology and therapy.Crit. Rev. Clin. Lab. Sci. 43, 143

42 Kratz, F. (2008) Albumin as a drug carrier: design of prodrugs, drugconjugates and nanoparticles. J. Control. Release 132, 171–183

43 Hawkins, M.J. et al. (2008) Protein nanoparticles as drug carriers inclinical medicine. Adv. Drug Deliv. Rev. 60, 876–885

44 Haley, B. and Frenkel, E. (2008) Nanoparticles for drug delivery incancer treatment. Urol. Oncol. 26, 57–64

45 Galley, H.F. and Webster, N.R. (2004) Physiology of the endothelium.Br. J. Anaesth. 93, 105–113

46 Desai, N. et al. (2006) Increased antitumor activity, intratumorpaclitaxel concentrations, and endothelial cell transport ofCremophor-free, albumin-bound paclitaxel, ABI-007, compared withCremophor-based paclitaxel. Clin. Cancer Res. 12, 1317–1324

47 Lu, W. et al. (2007) Brain delivery property and accelerated bloodclearance of cationic albumin conjugated PEGylated nanoparticle. J.Control. Release 118, 38–53

48 Zensi, A. et al. (2009) Albumin nanoparticles targeted with Apo E enterthe CNS by transcytosis and are delivered to neurones. J. Control.Release 137, 78–86

49 Lundberg, B.B. et al. (2004) Cellular association and cytotoxicity ofanti-CD74-targeted lipid drug-carriers in B lymphoma cells. J. Control.Release 94, 155–161

854

7

RQ2

855

856

857

858

859

860

861

862

863

864

865

866

867

868

869

870

871

872

873

874

875

876

877

878

879

880

881

882

883

884

885

886

887

888

889

890

891

892

893

894

895

Review Trends in Pharmacological Sciences Vol.xxx No.x

TIPS 737 1–8

50 Oh, S. et al. (2009) Synthesis and anti-cancer activity of covalentconjugates of artemisinin and a transferrin-receptor targetingpeptide. Cancer Lett. 274, 33–39

51 Kim, D.H. and Martin, D.C. (2006) Sustained release ofdexamethasone from hydrophilic matrices using PLGAnanoparticles for neural drug delivery. Biomaterials 27, 3031–

303752 Zhang, L. et al. (2008) Tumor targeting of vincristine by mBAFF-

modified PEG liposomes in B lymphoma cells. Cancer Lett. 269, 26–3653 Jain, A. and Jain, S.K. (2008) In vitro and cell uptake studies for

targeting of ligand anchored nanoparticles for colon tumors. Eur. J.Pharm. Sci. 35, 404–416

54 Wu, D.Q. et al. (2009) Galactosylated fluorescent labeled micelles as aliver targeting drug carrier. Biomaterials 30, 1363–1371

55 Yoo, H.S. et al. (2000) In vitro and in vivo anti-tumor activities ofnanoparticles based on doxorubicin-PLGA conjugates. J. Control.Release 68, 419–431

56 Kubek, M.J. et al. (2009) Attenuation of kindled seizures by intranasaldelivery of neuropeptide-loaded nanoparticles. Neurotherapeutics 6,359–371

57 Feng, S.S. et al. (2009) Poly(lactide)-vitamin E derivative/montmorillonite nanoparticle formulations for the oral delivery ofDocetaxel. Biomaterials 30, 3297–3306

UN

CO

RR

EC

8

OO

F

58 Juillerat-Jeanneret, L. (2008) The targeted delivery of cancer drugsacross the blood–brain barrier: chemical modifications of drugs or drug-nanoparticles? Drug Discov Today 13, 1099–1106

59 Swan, E.E.L. et al. (2008) Inner ear drug delivery for auditoryapplications. Adv. Drug Deliv. Rev. 60, 1583–1599

60 Liu, J. et al. (2008) Solid lipid nanoparticles for pulmonary delivery ofinsulin. Int. J. Pharm. 356, 333–344

61 Hajos, F. et al. (2008) Inhalable liposomal formulation for vasoactiveintestinal peptide. Int. J. Pharm. 357, 286–294

62 Vyas, S.P. et al. (2005) Aerosolized liposome-based delivery ofamphotericin B to alveolar macrophages. Int. J. Pharm. 296, 12–25

63 Tom, R.T. et al. (2004) Ciprofloxacin-protected gold nanoparticles.Langmuir 20, 1909–1914

64 Pandey, R. et al. (2003) Nanoparticle encapsulated antituberculardrugs as a potential oral drug delivery system against murinetuberculosis. Tuberculosis 83, 373–378

65 Moraes, C.M. et al. (2009) Benzocaine loaded biodegradable poly-(D,L-lactide-co-glycolide)nanocapsules: factorial designand characterization.Mater. Sci. Eng. B (in press) doi:10.1016/j.mseb.2009.06.011.

66 Manjunath, K. and Venkateswarlu, V. (2005) Pharmacokinetics, tissuedistribution and bioavailability of clozapine solid lipid nanoparticlesafter intravenous and intraduodenal administration. J. Control.Release 107, 215–228

TED

P

896


Recommended