Accepted Manuscript Title: Nanomedicine in veterinary oncology Author: Tzu-yin Lin, Carlos O. Rodriguez Jr, Yuanpei Li PII: DOI: Reference:

S1090-0233(15)00076-3 http://dx.doi.org/doi:10.1016/j.tvjl.2015.02.015 YTVJL 4430

To appear in:

The Veterinary Journal

Accepted date:

11-2-2015

Please cite this article as: Tzu-yin Lin, Carlos O. Rodriguez Jr, Yuanpei Li, Nanomedicine in veterinary oncology, The Veterinary Journal (2015), http://dx.doi.org/doi:10.1016/j.tvjl.2015.02.015. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20

Review

21



Nanomedicine in veterinary oncology Tzu-yin Lin a,*, Carlos O. Rodriguez Jr b, Yuanpei Li c a

Department of Internal Medicine, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA b Department of Veterinary Surgical and Radiological Sciences, School of Veterinary Medicine, University of California-Davis, Davis, CA 95616, USA c Department of Biochemistry and Molecular Biology, School of medicine, University of California-Davis, Sacramento, CA 95817, USA

* Corresponding author. Tel.: +1 916 7035081. E-mail address: [email protected] (T.-Y. Lin). Highlights

22 23

chemistry, biology and material sciences. 

24 25

Nanoparticle-based agents are currently under intensive investigation for cancer treatment in humans and animals.



26 27

Nanomedicine is an interdisciplinary field that combines medicine, engineering,

Nanoparticle-based agents can overcome several limitations associated with conventional oncology protocols.



Nanoparticles can be applied for cancer diagnosis and imaging.

28

Page 1 of 32

29

Abstract

30

Nanomedicine is an interdisciplinary field that combines medicine, engineering,

31

chemistry, biology and material sciences to improve disease management and can be especially

32

valuable in oncology. Nanoparticle-based agents that possess functions such as tumor targeting,

33

imaging and therapy are currently under intensive investigation. This review introduces the basic

34

concept of nanomedicine and the classification of nanoparticles. Because of their favorable

35

pharmacokinetics, tumor targeting properties, and resulting superior efficacy and toxicity profiles,

36

nanoparticle-based agents can overcome several limitations associated with conventional

37

diagnostic and therapeutic protocols in veterinary oncology.

38 39

The two most important tumor targeting mechanisms (passive and active tumor targeting)

40

and their dominating factors (i.e. shape, charge, size and nanoparticle surface display) are

41

discussed. The review summarizes published clinical and preclinical studies that utilize different

42

nanoformulations in veterinary oncology, as well as the application of nanoparticles for cancer

43

diagnosis and imaging. The toxicology of various nanoformulations is also considered. Given the

44

benefits of nanoformulations demonstrated in human medicine, nanoformulated drugs are likely

45

to gain more traction in veterinary oncology.

46 47

Keywords: Nanomedicine; Nanoparticles; Chemotherapy; Drug delivery; Oncology; Veterinary

48

Page 2 of 32

49 50

Introduction Nanotechnology is an emerging field that has shown great promise in the development of

51

novel diagnostic, imaging and therapeutic agents for a variety of diseases, including cancer

52

(Davis et al., 2008). It exploits the improved and often novel physical, chemical and biological

53

properties of materials on a nanometer scale. Nanomedicine is defined as the application of

54

nanotechnology to medical diagnosis, therapy and prevention.

55 56

Nanoformulations seek to overcome several limitations of conventional drugs, including

57

toxicity, poor water solubility, instability (e.g. small interfering RNA, or siRNA) and

58

pharmacokinetic (PK) properties, and may also contribute to the advancement of personalized

59

medicine and to the customization of healthcare (Eifler and Thaxton, 2011). Taking advantage of

60

versatile payloads, favorable PKs, unique tumor targeting properties with both passive and active

61

mechanisms, and an overall superior efficacy and toxicity profile, these nanoscale ‘theranostic’

62

(therapeutic-diagnostic, i.e. combining therapeutic and diagnostic purposes) formulations represent

63

potential breakthroughs for cancer therapy and have created a new field known as ‘cancer

64

nanomedicine’ (Chow and Ho, 2013).

65 66

Companion animals, such as cats and dogs, spontaneously develop various types of

67

cancers, such as oral squamous cell carcinoma (SCC), mammary carcinoma, osteosarcoma (OSA)

68

and transitional cell carcinomas, which closely resemble cancers in humans (Rowell et al., 2011).

69

Consequently, spontaneous cancers in cats and dogs have been proposed as the best animal

70

models for human cancers and have been used in preclinical studies for novel drug development,

Page 3 of 32

71

including nanoformulated drugs or imaging probes (De Vico et al., 2005; Withrow and Wilkins,

72

2010; Rowell et al., 2011).

73 74 75

This review focuses on nanoformulations that have been reported in preclinical studies using companion animals or in early phase clinical trials in veterinary medicine.

76 77

Nanoparticle classification and tumor targeting properties

78

Composition of nanoparticles

79

Nanoparticles can be categorized as inorganic or ‘solid’ (gold, iron oxide, quantum dots

80

and carbon nanotubes), or as organic or ‘soft’ (liposomes, dendrimers, polymeric micelles, and

81

protein aggregates (Yu et al., 2012; Bao et al., 2013; Cheng et al., 2014). Each nanoparticle

82

category has distinct advantages and limitations; for instance, quantum dots and iron oxide

83

particles have well known fluorescence imaging capability and magnetic resonance imaging

84

(MRI) contrast properties, respectively, but are limited as drug delivery vehicles (Lovell et al.,

85

2011; Li et al., 2014). Conversely, liposomes and polymeric micelles are used clinically for drug

86

delivery, but offer limited applications as imaging agents.

87 88

Novel nanomedicine platforms can be developed that synthesize hybrid nanoparticles.

89

Specifically, these synthetic theranostic nanoparticles merge the therapeutic potential of the

90

polymeric soft nanoparticle domains with the diagnostic properties of inorganic solid

91

nanoparticles. Most recently, a few novel organic theranostic nanoplatforms have been touted for

92

their potential applications in optical imaging, MRI, positron emission tomography (PET),

Page 4 of 32

93

chemotherapy, photodynamic therapy (PDT) and photothermal therapy (Lovell et al., 2011; Li et

94

al., 2014).

95 96 97

Factors associated with passive tumor targeting The tumor vasculature and lymphatic vessels are known to be leaky to macromolecules.

98

Thus, nanoparticles can preferentially accumulate in tumors via enhanced permeability and

99

retention (EPR) effects (Matsumura and Maeda, 1986) (Fig. 1). Size, surface charge and shape

100

dictate the interaction of nanoparticles in living subjects. These nanoparticle-specific properties

101

will affect their PK, biodistribution and diffusivity, and consequently determine their in vivo

102

efficacy and toxicity profiles.

103 104

Nanoparticle size affects the rate of nanoparticle intratumoral deposition through the EPR

105

effect and therapeutic efficacy. The optimal nanoparticle size for passive tumor targeting is ~10-

106

100 nm (Davis et al., 2008). Hydrophilic components, such as polyethylene glycol (PEG), have

107

been used to coat the surface of nanoparticles to minimize their interaction with blood proteins

108

and reduce their subsequent sequestration by macrophages (Gref et al., 1994; Zahr et al., 2006;

109

Schipper et al., 2009). Unfortunately, the generation of anti-PEG IgM results in accelerated

110

blood clearance (ABC) and decreased liposomal drug circulation time (Suzuki et al., 2012; Abu

111

Lila et al., 2013) (Table 1, liposomal topotecan in dogs) with repeated PEGylated liposome

112

administration.

113 114 115

The surface charge (i.e. positive, neutral or negative) and density also need to be optimized to prolong the blood circulation time, minimize non-specific clearance and prevent

Page 5 of 32

116

loss to undesired locations. In addition, the shape of nanoparticles affects their blood circulation,

117

ability to marginate and binding affinity, and therefore the rate of tumor deposition and

118

therapeutic efficacy; for example, rods and hollow cubes enter tumors more readily than discs or

119

spheres (Wang et al., 2013b).

120 121

Active cancer targeting strategy

122

Active targeting is more attractive than passive EPR because of improved efficiency,

123

specific delivery of more therapeutic drugs/probes to target sites and the potential for

124

individualized treatment. As shown in Fig. 1, taking advantage of specific cancer cell surface

125

receptors (e.g. folate, transferrin, asialoglycoprotein, integrins, epidermal growth factor

126

receptors, CD44) and unique tumor microenvironment signaling molecules (e.g. vascular

127

endothelial growth factor, matrix metalloproteinases, αvβ3 integrin), a broad variety of ligands

128

(e.g. antibodies, single-chain Fv fragments, peptides, small molecules, aptamers) can be bound to

129

the surface of nanoparticles for cancer-targeting therapy (Zhang et al., 2007; Dhar et al., 2008;

130

McCarron et al., 2008; Lu et al., 2009). Compared to the EPR effect alone, these ‘active’

131

targeting methods enhance delivery, allow for deeper tumor penetration, and prolong drug

132

retention within both the blood and the tumor, resulting in superior anti-cancer efficacy,

133

specificity and biodistribution.

134 135

Our laboratory has identified a urinary bladder cancer specific peptide, PLZ4, which

136

specifically recognizes dog and human neoplastic, but not normal or inflamed urothelial cells,

137

fibroblasts or white blood cells. When PLZ4 is displayed on the surface of micelles by the self-

138

assembly of telodendrimers, these ‘active bladder cancer targeting micelles’ are considered to be

Page 6 of 32

139

theranostic because they can deliver chemotherapeutic drugs (doxorubicin or paclitaxel) and

140

fluorescence dyes to bladder cancer cells (Fig. 2). These micelles enhance the anti-cancer

141

efficacy in a bladder xenograft mouse model.

142 143 144

Nanoparticle-based drug delivery systems for veterinary oncology Nanoparticles have been used to deliver chemotherapeutic drugs, small molecule

145

inhibitors and cytokines to tumor sites. Different formulations may enhance the therapeutic index,

146

increase the maximally tolerated dose (MTD), exhibit preferable PKs or improve the toxicity

147

profile of existing drugs. The nanoparticle approach has also offered a second chance for drugs

148

that are effective but toxic, poorly water soluble drugs, or unstable molecules (e.g. siRNA) for

149

applications in veterinary clinical studies. Nanocarriers and their payloads, delivery routes, and

150

neoplastic targets evaluated in companion animals are summarized in Table 1.

151 152

Paclitaxel nanoformulations

153

The first US Food and Drug Administration (FDA) approved nanoformulation in

154

veterinary oncology is Paccal Vet, which consists of paclitaxel (PTX)-loaded micelles. It

155

received conditional approval in 2014 for the treatment of canine mammary carcinoma and

156

squamous cell carcinoma. Although paclitaxel is effective against a broad range of human

157

cancers, its use in veterinary medicine has been hampered by hypersensitivity reactions to the

158

PTX carrier, cremaphor. Paccal Vet also improved the overall response rate and biological

159

observed response rate of canine malignant mast cell tumors (MCTs) when compared to

160

lomustine (Vail et al., 2012). In a subsequent study, 59% complete or partial response rates were

161

documented in 29 grade II-III canine MCTs (Rivera et al., 2013).

Page 7 of 32

162 163 164

Doxorubicin nanoformulations Liposomes have been used to deliver chemotherapeutic drugs (doxorubicin; DOX),

165

cytokines (interleukin 2, IL-2), small molecule inhibitors and immune active reagents (muramyl

166

tripeptide phosphatidyl ethanolamine, MTP) in veterinary oncology (MacEwen et al., 1989;

167

Khanna et al., 1997; Poirier et al., 2002; Hauck et al., 2006). DOX-encapsulated (PEGylated or

168

hybrid) liposomes have been investigated in dogs and cats for their clinical efficacy against

169

sarcomas (Poirier et al., 2002; Sorenmo et al., 2007; Kleiter et al., 2010), carcinomas (Hauck et

170

al., 2006) and round cell tumors (Kisseberth et al., 1995; Vail et al., 1998; Stettner et al., 2005).

171

Doxil is the first FDA-approved liposomal DOX in human medicine and has also been studied in

172

veterinary oncology (Barenholz, 2012). In the clinical trial of Doxil, 51 tumor-bearing dogs were

173

enrolled and a 25.5% overall response rate was observed (Vail et al., 1998). Interestingly,

174

intraperitoneal administration of Doxil failed to demonstrate improvement in overall survival or

175

prevention of peritoneal recurrence when compared to conventionally formulated and

176

administered free DOX given post-operatively to dogs with splenic hemangiosarcoma (HSA)

177

(Sorenmo et al., 2007).

178 179

Doxil (1 mg/kg) has been used in cats with vaccine-associated sarcoma (VAS), with a

180

39% overall response rate and a median progression time of 84 days. This was similar to cats

181

receiving free DOX (Poirier et al., 2002). Overall, Doxil was well-tolerated and had less

182

cardiotoxicity, despite similar efficacy. However, despite this favorable toxicity profile, cost has

183

prevented its widespread use in veterinary medicine.

184

Page 8 of 32

185 186

Immunomodulator nanoformulations Liposomes have been used to deliver the immunomodulator MTP for various cancers in

187

dogs (OSA, HSA, malignant melanoma and mammary carcinoma) and cats (mammary

188

carcinoma) (MacEwen et al., 1989, 1999; Fox et al., 1995; Kurzman et al., 1995; Vail et al.,

189

1995). Dogs with OSA and splenic HSA receiving liposomal MTP had significantly prolonged

190

disease-free survival, overall survival and/or prolonged time to relapse (Kleinerman et al., 1995;

191

Kurzman et al., 1995; Vail et al., 1995). However, liposomal MTP failed to prolong post-surgical

192

survival rates in dogs with oral malignant melanoma or mammary carcinoma (MacEwen et al.,

193

1999; Teske et al., 1998). There is still a strong interest in the use of liposomal MTP in the

194

treatment of human OSA and the formulation is currently in early clinical trials in humans in

195

Europe. The drug has not received FDA approval in the United States and hence its availability

196

is restricted.

197 198 199

Nanoparticle-mediated photodynamic and photothermal therapy Nanoparticle-mediated photodynamic therapy (PDT) and photothermal therapy (PTT) are

200

interesting alternative approaches for cancer treatment. First and second generation

201

photosensitizers have poor water solubility, equivocal cancer selectivity and often accumulate in

202

the skin for prolonged periods of time after therapy, which leads to photosensitivity and sunburns.

203

Nanoformulated photosensitizers are currently considered as third generation photosensitizers

204

and presumably have better tumor targeting property.

205 206

A preliminary trial in feline cutaneous SCC using a liposomal formulation of the

207

lipophilic photosensitizer meta-(tetrahydroxyphenyl) chlorin (m-THPC)-mediated PDT resulted

Page 9 of 32

208

in a 100% complete response rate, a 75% overall 1-year control rate and a 20% recurrence rate.

209

No obvious systemic toxicity was noted but, importantly, untoward local skin reactions occurred

210

in only 15% of the patients (Buchholz et al., 2007).

211 212

An orthotopic canine transmissible venereal tumor (cTVT) brain tumor model has been

213

tested for gold nanoshell-assisted PTT. After infusion, the nanoshells specifically accumulated in

214

the intracranial cTVT through the disrupted blood-brain barrier. Intratumoral laser illumination

215

thermally ablated the cTVTs (average temperature 65.8 ± 4.1 °C). Importantly, the temperature

216

achieved in the vehicle-treated control dog (53.1 °C) resulted in minimal normal tissue damage

217

due the lack of nanoshell accumulation (Schwartz et al., 2009). Currently, The Ohio State

218

University has an open enrolment for dogs with solid tumors for a phase I study on gold

219

nanorods-mediated PTT (Table 2).

220 221 222

Alternative administrating route for nanoparticles As well as intravenous (IV) administration, nanoparticles have also been given via

223

intrathecal (Kitamura et al., 1996), intratumoral (Kitamura et al., 1996), inhalational (Khanna et

224

al., 1997) and intravesicular (Lu et al., 2011) routes. Canine transitional cell carcinoma of the

225

urinary bladder has been used to evaluate intravesicular administration of PTX-loaded gelatin-

226

based nanoparticles with minimal systemic absorption and toxicity (Lu et al., 2011). Comparing

227

neoplastic to normal urothelial cells using PTX-gelatin nanoparticle, a differential uptake as high

228

as 360-fold at the same tissue depth was observed (Lu et al., 2011).

229

Page 10 of 32

230

A carrier that can penetrate the blood-brain barrier is usually required for targeting brain

231

tumor cells via systemic administration; however, Dickinson et al. (2010) bypassed this

232

requirement by directly injecting liposomal CP-11 (a topoisomerase inhibitor)/gadolinium (Gd)

233

into canine spontaneous gliomas. Drug distribution and tumor response was monitored by MRI;

234

5/9 dogs exhibited decreased tumor volume (40-88%) (Dickinson et al., 2010). To date, no

235

liposomal formulations administered IV to dogs with brain tumors have been studied in a similar

236

manner. It is tempting to speculate that nanoparticles could be created to penetrate the blood-

237

brain barrier and deliver their theranostic payload to brain tumors, thus leading to similar or

238

better results without the necessity for technically demanding intrathecal injections. However,

239

further experiments are necessary to demonstrate feasibility.

240 241 242

Nanoparticles for cancer diagnosis and imaging applications Rapid advances in the nanoprobe field have created a novel targeted non-invasive method

243

to evaluate both the tumor and its microenvironment. These advancements further enable

244

scientists to detect, control biodistribution and monitor treatment in real time (Shin et al., 2013).

245

However, there are very few limited studies on the application of nanoparticles for imaging of

246

cancers in companion animals.

247 248

Several inorganic nanoparticles have been tested in veterinary medicine. In client-owned

249

dogs with spontaneous thyroid carcinoma and OSA, gum arabic-stabilized gold nanocrystals

250

have been injected intratumorally as contrast agents. Computed tomography (CT) images

251

showed effective accumulation at the tumor sites with a contrast enhancement of 12 δ-HU

252

(Chanda et al., 2014). Similarly, Technetium-99m-labelled liposomes provided scintigraphic

Page 11 of 32

253

imaging with a four-fold image enhancement of feline sarcomas undergoing tumor hyperthermia

254

(Matteucci et al., 2000). These studies support the potential use of nanoformulations to detect

255

tumors, monitor drug delivery, control drug release and assess treatment outcomes.

256 257

Canine spontaneous glioma has been used as a model for drug delivery, imaging, and

258

therapy due to its striking similarities to human glioma. It is a challenge to treat glioma

259

systemically due to the blood-brain barrier. Convection-enhanced delivery (CED) of therapeutic

260

agents has circumvented the blood-brain barrier by allowing direct infusion of test articles

261

directly into tumors via one to multiple catheters. Nanotechnology offers a multifunctional

262

platform allowing real-time imaging and monitoring of drug delivery, while sequentially

263

accurately evaluating therapeutic efficacy. As mentioned above, through CED, Dickinson et al.

264

(2010) co-infused CPT-11/Gd-encapsulated liposomes into dogs with spontaneous grade III

265

astrocytoma and used concurrent real-time MRI visualization. The distribution, location and

266

leakage of the infusate could be observed on T1-weighted images; coupling treatment with

267

imaging (theranostic) helped to optimize infusion parameters and the interpretation of outcomes

268

(Dickinson et al., 2010).

269 270

Our group has studied the biodistribution of indodicarbocyanines (DiD, a near infrared,

271

NIR, fluorescent dye)/PTX co-loaded PLZ4-micelles (PNMs) via optical imaging in a canine

272

urinary bladder cancer orthotopic mouse model. PNMs exhibited significantly higher

273

accumulation in tumors than non-targeted micelles as assessed by NIR fluorescent signals (Lin et

274

al., 2012). This selectivity toward cancer suggested that our PNMs could carry and deliver

Page 12 of 32

275

chemotherapeutic drugs to tumor sites and could be used for intravesicular tumor detection in

276

dogs. This possibility is currently being assessed in a clinical trial in dogs with urinary bladders.

277 278

Several other nanomaterials have been injected into dogs to test their potential for

279

diagnosing tumors or for mapping tumor draining lymph nodes. For example, cetuximab-

280

conjugated magnetic iron-oxide nanoparticles and liposome-encapsulated Gd were infused in

281

dog brains to evaluate toxicity and their potential application in glioma diagnosis and treatment

282

(Dickinson et al., 2008; Platt et al., 2012). Lymph node metastases are the hallmark for various

283

malignant cancers. Lymphadenectomy, along with primary tumor resection, may achieve better

284

long-term cancer control and prevent local recurrences. Iodized oil emulsion has been injected

285

into the gastric submucosa so that draining lymph nodes could be identified by CT and

286

lymphangiography (Lim et al., 2012). Furthermore, lymphotropic iodinated nanoparticles have

287

been injected subcutaneously near oral malignant melanomas to detect cancerous lymph nodes

288

(Wisner et al., 1996).

289 290 291

Toxicology Nanoparticle formulation not only improves drug delivery but also largely changes the

292

PKs of chemotherapeutic drugs, resulting in reduced toxicity and side effects (De Jong and Borm,

293

2008). Some effective but poor water soluble chemotherapeutic drugs, such as PTX, can now be

294

used in veterinary oncology. Because of the emerging recognition of companion animals with

295

spontaneous cancers as animal models with great potential for human oncology research, many

296

anti-cancer nanoformulations have been tested in dogs, rabbits and cats to predict safety issues

297

and to determine the recommended starting dose in human clinical trials. Although most

Page 13 of 32

298

molecules are not aimed at veterinary species, those preclinical studies offer a wealth of

299

information regarding potential toxicity in animals.

300 301

Toxicity associated with doxorubicin nanoformulations

302

DOX-containing liposomes were studied for their toxicity in dogs and rabbits in the late

303

1990s. DOX is an effective chemotherapeutic drug, but cardiotoxicity limits its use in dogs and

304

humans. Dogs treated with Doxil showed no evidence of cardiotoxicity clinically or

305

pathologically, while dogs treated with free DOX exhibited marked cardiotoxicity. Similarly,

306

rabbits receiving Doxil had lower evidence of cardiotoxicity (16%) compared with free DOX

307

treated rabbits (67%) (Working et al., 1999). Findings in humans reflect these preclinical

308

findings; Doxil effectively minimizes cardiomyopathy even when patients receive high

309

cumulative doses (Gabizon et al., 2004; Uyar et al., 2004). Although the Doxil formulation

310

prolonged DOX circulating time in dogs and changed its biodistribution, resulting in decreased

311

cardiotoxicity, there was a higher incidence of skin toxicity, such as palmar-plantar

312

erythrodysesthesias (PPE), the dose-limiting event in dogs and humans. Pyridoxine (vitamin B6)

313

delays and decreases the development and severity of PPE in lymphoma-bearing dogs receiving

314

Doxil (Vail et al., 1998). In comparison, myelosuppresion with free DOX treatment is usually the

315

dose-limiting toxicity (Vail et al., 1997).

316 317

Doxil has also been evaluated in cats with vaccine-associated sarcoma. Delayed

318

nephrotoxicity and cutaneous toxicity occurred in 23% and 22% of treated cats, respectively

319

(Poirier et al., 2002). Overall, Doxil has been considered well-tolerated in cancer veterinary

320

patients and has demonstrated decreased cardiotoxicity with manageable cutaneous toxicity.

Page 14 of 32

321 322

Another albumin-binding DOX prodrug, the 6-maleimidocaproyl hydrazone derivative of

323

doxorubicin (DOXO-EMCH), was tested in dogs to determine the recommended starting dose in

324

humans and to evaluate potential toxicities. From the time of injection up to 3 h, dogs showed

325

dose-dependent allergic reactions, along with clinical signs of extensive skin redness, swelling,

326

and salivation. At the high dose, some animals also developed post-injection motor hypoactivity,

327

hair loss and dermatitis (Kratz et al., 2007). Of note, the free DOX MTD is approximately 2.25-

328

2.5 mg/kg in dogs, while DOXO-EMCH can double the dose of the free DOX MTD (Kratz et al.,

329

2007).

330 331

Toxicity associated with plaxitel nanoformulations

332

PTX, a mitotic inhibitor, is a potent anti-cancer drug in humans; however, despite

333

aggressive premedication in dogs and cats, cremophor EL-induced side effects largely limits its

334

veterinary use (Poirier et al., 2004; Kim et al., 2014). Nanotechnology has enabled the

335

production of a canine-tolerated PTX (Paccal Vet; PTX-loaded micelles; MTD 150 mg/m2).

336 337

In the first randomized trial of Paccal Vet in dogs with high grade MCTs, the most

338

common adverse event was grade 3-4 neutropenia, followed by the gastrointestinal toxicity

339

(emesis, anorexia and diarrhea) (Vail et al., 2012). Increased liver enzymes were noted, but to a

340

less extent than in the control dogs that received lomustine, a standard chemotherapeutic drug.

341

While 33% of dogs receiving lomustine were withdrawn due to clinically relevant hepatotoxicity,

342

only 2% of dogs receiving Paccal Vet did so (Vail et al., 2012). These findings were confirmed

Page 15 of 32

343

in later trials using dogs with high grade solid tumors and MCTs (Rivera et al., 2013; von Euler

344

et al., 2013).

345 346

Nanoparticulate PTX in saline (Crititax, CTI52010) has an MTD of 120 mg/m2 in dogs

347

and also causes grade 4 neutropenia and grade 1-2 gastrointestinal toxicity at higher doses

348

(Axiak et al., 2011). Intraparenchymal microsphere-PTX (Paclimer) delivery to the brain is not

349

associated with any systemic toxicity or myelosuppression. Although no neurotoxicity was found,

350

local wound infection and brain abscess were present on occasions (Pradilla et al., 2006). These

351

results were as expected; only local exposure occurred without systemic absorption.

352 353

Since dogs are more sensitive to the development of anaphylaxis in response to treatment

354

with various PTX formulations, they have also been used to evaluate allergic reactions after

355

systemic administration of PTX microemulsions (Wang et al., 2011). Compared to dogs

356

receiving PTX (in cremophor EL/ethanol), dogs receiving PTX microemulsion exhibited

357

significantly less severe allergic reactions. Premedication with prednisone, diphenhydramine,

358

cimetidine and dexamethasone significantly inhibited allergic reactions induced by PTX and

359

PTX microemulsion in dogs. The group of dogs receiving PTX microemulsion with

360

premedication exhibited the least toxicity (Wang et al., 2011).

361 362

Shi et al. (2013) studied the toxicity and PKs of docetaxel-loaded arginine-stabilized

363

mPEG-PDLLA polymeric micelles in dogs and showed that this micelle formulation also

364

improved the acute toxicity that occurred with free docetaxel injection. Taken together, PTX

365

nanoformulations have superior toxicity profiles when to conventional, poorly water soluble

Page 16 of 32

366

PTX. Most adverse effects to the nanoformulations were transient, clinically silent, and

367

manageable.

368 369 370

Toxicity of other chemotherapeutic drug nanoformulation Liposomal curcumin causes reversible hematuria, while higher doses (40 mg/kg) caused

371

irreversible acute hemolysis accompanied by hematuria. Hemolysis was more likely related to

372

oxidative damage from the drug (Helson et al., 2012). CED CPT-11 liposome induces mild

373

transient proprioceptive deficits, local hemorrhage and perivascular inflammation following

374

direct intraparenchymal delivery (Dickinson et al., 2008).

375 376

Interestingly, the toxicity profiles of the same nanoformulation may vary across species.

377

For instance, in mice, liposome encapsulated vincristine (an organic nanoparticle) caused

378

significant less toxicity when compared to free vincristine following intravascular injection,

379

while dogs exhibited comparable toxicity between those two molecules (gastrointestinal toxicity

380

was the dose-limiting factor when given as a single high-dose) (Kanter et al., 1994). Toxicology

381

studies in veterinary medicine are lacking, especially for those inorganic nanomaterials.

382 383

Conclusions

384

The field of nanotechnology is growing rapidly. The technology has enabled the clinical

385

use of several chemotherapeutic drugs that were originally too toxic, too unstable, or difficult to

386

formulate. Veterinary medicine is poised to take advantage of these advances. In humans,

387

nanoparticle drug formulation has shown clinically superior tumor targeting ability, favorable

388

patient PK/biodistribution, and an overall improved toxicity profile. There are an increasing

Page 17 of 32

389

number of nanoformulations receiving approval from regulatory agencies in human medicine,

390

while the first paclitaxel nanoformulation was approved by the FDA for companion animal use

391

in 2014. Despite these advances, most formulations are still in their very early development.

392

More and different formulations are expected to be tested and approved for clinical use and for

393

development as theranostic nanoformulations, which will allow for convenient detection,

394

treatment and imaging follow-up of tumors in a single platform. Most importantly, advances in

395

targeting of tumors or tumor environments will further contribute to the development of

396

individualized medicine. All of these steps will eventually influence cancer treatment in human

397

and veterinary medicine.

398 399 400 401

Conflict of interest statement None of the authors of this paper has a financial or personal relationship with other people or organisations that could inappropriately influence or bias the content of the paper.

402 403

References

404 405 406 407 408 409 410 411 412 413 414 415 416 417 418

Abu Lila, A.S., Kiwada, H., Ishida, T., 2013. The accelerated blood clearance (ABC) phenomenon: Clinical challenge and approaches to manage. Journal of Controlled Release 172, 38-47. Amantea, M., Newman, M.S., Sullivan, T.M., Forrest, A., Working, P.K., 1999. Relationship of dose intensity to the induction of palmar-plantar erythrodysesthia by pegylated liposomal doxorubicin in dogs. Human and Experimental Toxicology 18, 17-26. Axiak, S.M., Selting, K.A., Decedue, C.J., Henry, C.J., Tate, D., Howell, J., Bilof, K.J., Kim, D.Y., 2011. Phase I dose escalation safety study of nanoparticulate paclitaxel (CTI 52010) in normal dogs. International Journal of Nanomedicine 6, 2205-2212. Barenholz, Y., 2012. Doxil (R) the first FDA-approved nano-drug: Lessons learned. Journal of Controlled Release 160, 117-134.

Page 18 of 32

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

Buchholz, J., Wergin, M., Walt, H., Grafe, S., Bley, C.R., Kaser-Hotz, B., 2007. Photodynamic therapy of feline cutaneous squamous cell carcinoma using a newly developed liposomal photosensitizer: Preliminary results concerning drug safety and efficacy. Journal of Veterinary Internal Medicine 21, 770-775. Bao, G., Mitragotri, S., Tong, S., 2013. Multifunctional nanoparticles for drug delivery and molecular imaging. Annual Review of Biomedical Engineering 15, 253-282. Chanda, N., Upendran, A., Boote, E.J., Zambre, A., Axiak, S., Selting, K., Katti, K.V., Leevy, W.M., Afrasiabi, Z., Vimal, J., et al., 2014. Gold nanoparticle based X-ray contrast agent for tumor imaging in mice and dog: A potential nano-platform for computer tomography theranostics. Journal of Biomedical Nanotechnology 10, 383-392. Cheng, Y., Morshed, R.A., Auffinger, B., Tobias, A.L., Lesniak, M.S., 2014. Multifunctional nanoparticles for brain tumor imaging and therapy. Advanced Drug Delivery Reviews 66, 42-57. Chow, E.K., Ho, D., 2013. Cancer nanomedicine: From drug delivery to imaging. Science Translational Medicine 5, 216. Davis, M.E., Chen, Z.G., Shin, D.M., 2008. Nanoparticle therapeutics: An emerging treatment modality for cancer. Nature Reviews Drug Discovery 7, 771-782. De Jong, W.H., Borm, P.J., 2008. Drug delivery and nanoparticles: Applications and hazards. International Journal of Nanomedicine 3, 133-149. De Vico, G., Maiolino, P., Restucci, B., Passantino, A., 2005. Spontaneous tumours of pet dog as models for human cancers: Searching for adequate guidelines. Rivista di Biologia 98, 279-296. Dhar, S., Gu, F.X., Langer, R., Farokhzad, O.C., Lippard, S.J., 2008. Targeted delivery of cisplatin to prostate cancer cells by aptamer functionalized Pt(IV) prodrug-PLGA-PEG nanoparticles. Proceedings of the National Academy of Sciences of the United States of America 105, 17356-17361. Dickinson, P.J., LeCouteur, R.A., Higgins, R.J., Bringas, J.R., Larson, R.F., Yamashita, Y., Krauze, M.T., Forsayeth, J., Noble, C.O., Drummond, D.C., et al., 2010. Canine spontaneous glioma: A translational model system for convection-enhanced delivery. Neuro-oncology 12, 928-940. Dickinson, P.J., LeCouteur, R.A., Higgins, R.J., Bringas, J.R., Roberts, B., Larson, R.F., Yamashita, Y., Krauze, M., Noble, C.O., Drummond, D., et al., 2008. Canine model of convection-enhanced delivery of liposomes containing CPT-11 monitored with realtime magnetic resonance imaging: Laboratory investigation. Journal of Neurosurgery 108, 989-998.

Page 19 of 32

465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509

Dow, S., Elmslie, R., Kurzman, I., MacEwen, G., Pericle, F., Liggitt, D., 2005. Phase I study of liposome-DNA complexes encoding the interleukin-2 gene in dogs with osteosarcoma lung metastases. Human Gene Therapy 16, 937-946. Eifler, A.C., Thaxton, C.S., 2011. Nanoparticle therapeutics: FDA approval, clinical trials, regulatory pathways, and case study. Methods in Molecular Biology 726, 325-338. Fox, L.E., MacEwen, E.G., Kurzman, I.D., Dubielzig, R.R., Helfand, S.C., Vail, D.M., Kisseberth, W., London, C., Madewell, B.R., Rodriguez, C.O., Jr., 1995. Liposomeencapsulated muramyl tripeptide phosphatidylethanolamine for the treatment of feline mammary adenocarcinoma - a multicenter randomized double-blind study. Cancer Biotherapy 10, 125-130. Fox, L.E., Toshach, K., Calderwood-Mays, M., Khokhar, A.R., Kubilis, P., Perez-Soler, R., MacEwen, E.G., 1999. Evaluation of toxicosis of liposome-encapsulated cis-bisneodecanoato-trans-R,R-1,2-diaminocyclohexane platinum (II) in clinically normal cats. American Journal of Veterinary Research 60, 257-263. Gabizon, A.A., Lyass, O., Berry, G.J., Wildgust, M., 2004. Cardiac safety of pegylated liposomal doxorubicin (Doxil/Caelyx) demonstrated by endomyocardial biopsy in patients with advanced malignancies. Cancer Investigation 22, 663-669. Gref, R., Minamitake, Y., Peracchia, M.T., Trubetskoy, V., Torchilin, V., Langer, R., 1994. Biodegradable long-circulating polymeric nanospheres. Science 263, 1600-1603. Hafeman, S., London, C., Elmslie, R., Dow, S., 2010. Evaluation of liposomal clodronate for treatment of malignant histiocytosis in dogs. Cancer Immunology Immunotherapy 59, 441-452. Hauck, M.L., LaRue, S.M., Petros, W.P., Poulson, J.M., Yu, D., Spasojevic, I., Pruitt, A.F., Klein, A., Case, B., Thrall, D.E., et al., 2006. Phase I trial of doxorubicin-containing low temperature sensitive liposomes in spontaneous canine tumors. Clinical Cancer Research 12, 4004-4010. Helson, L., Bolger, G., Majeed, M., Vcelar, B., Pucaj, K., Matabudul, D., 2012. Infusion pharmacokinetics of Lipocurc (liposomal curcumin) and its metabolite tetrahydrocurcumin in Beagle dogs. Anticancer Research 32, 4365-4370. Huttinger, C., Hirschberger, J., Jahnke, A., Kostlin, R., Brill, T., Plank, C., Kuchenhoff, H., Krieger, S., Schillinger, U., 2008. Neoadjuvant gene delivery of feline granulocytemacrophage colony-stimulating factor using magnetofection for the treatment of feline fibrosarcomas: A phase I trial. Journal of Gene Medicine 10, 655-667. Jahnke, A., Hirschberger, J., Fischer, C., Brill, T., Kostlin, R., Plank, C., Kuchenhoff, H., Krieger, S., Kamenica, K., Schillinger, U., 2007. Intra-tumoral gene delivery of feIL-2,

Page 20 of 32

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

feIFN-gamma and feGM-CSF using magnetofection as a neoadjuvant treatment option for feline fibrosarcomas: A phase-I study. Journal of Veterinary Medicine 54, 599-606. Kamstock, D., Guth, A., Elmslie, R., Kurzman, I., Liggitt, D., Coro, L., Fairman, J., Dow, S., 2006. Liposome-DNA complexes infused intravenously inhibit tumor angiogenesis and elicit antitumor activity in dogs with soft tissue sarcoma. Cancer Gene Therapy 13, 306317. Kanter, P.M., Klaich, G.M., Bullard, G.A., King, J.M., Bally, M.B., Mayer, L.D., 1994. Liposome encapsulated vincristine: Preclinical toxicologic and pharmacologic comparison with free vincristine and empty liposomes in mice, rats and dogs. AntiCancer Drugs 5, 579-590. Khanna, C., Anderson, P.M., Hasz, D.E., Katsanis, E., Neville, M., Klausner, J.S., 1997. Interleukin-2 liposome inhalation therapy is safe and effective for dogs with spontaneous pulmonary metastases. Cancer 79, 1409-1421. Kim, J., Doerr, M., Kitchell, B.E., 2014. Exploration of paclitaxel (Taxol) as a treatment for malignant tumors in cats: A descriptive case series. Journal of Feline Medicine and Surgery 17, 186-190. Kisseberth, W.C., MacEwen, E.G., Helfand, S.C., Vail, D.M., London, C.L., Keller, E., 1995. Response to liposome-encapsulated doxorubicin (TLC D-99) in a dog with myeloma. Journal of Veterinary Internal Medicine 9, 425-428. Kitamura, I., Kochi, M., Matsumoto, Y., Ueoka, R., Kuratsu, J., Ushio, Y., 1996. Intrathecal chemotherapy with 1,3-bis(2-chloroethyl)-1-nitrosourea encapsulated into hybrid liposomes for meningeal gliomatosis: An experimental study. Cancer Research 56, 3986-3992. Kiyokawa, H., Igawa, Y., Muraishi, O., Katsuyama, Y., Iizuka, K., Nishizawa, O., 1999. Distribution of doxorubicin in the bladder wall and regional lymph nodes after bladder submucosal injection of liposomal doxorubicin in the dog. Journal of Urology 161, 665667. Kleinerman, E.S., Gano, J.B., Johnston, D.A., Benjamin, R.S., Jaffe, N., 1995. Efficacy of liposomal muramyl tripeptide (CGP 19835A) in the treatment of relapsed osteosarcoma. American Journal of Clinical Oncology 18, 93-99. Kleiter, M., Tichy, A., Willmann, M., Pagitz, M., Wolfesberger, B., 2010. Concomitant liposomal doxorubicin and daily palliative radiotherapy in advanced feline soft tissue sarcomas. Veterinary Radiology and Ultrasound 51, 349-355. Kratz, F., Ehling, G., Kauffmann, H.M., Unger, C., 2007. Acute and repeat-dose toxicity studies of the (6-maleimidocaproyl)hydrazone derivative of doxorubicin (DOXO-EMCH), an

Page 21 of 32

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

albumin-binding prodrug of the anticancer agent doxorubicin. Human and Experimental Toxicology 26, 19-35. Kurzman, I.D., MacEwen, E.G., Rosenthal, R.C., Fox, L.E., Keller, E.T., Helfand, S.C., Vail, D.M., Dubielzig, R.R., Madewell, B.R., Rodriguez, C.O., Jr., et al., 1995. Adjuvant therapy for osteosarcoma in dogs: Results of randomized clinical trials using combined liposome-encapsulated muramyl tripeptide and cisplatin. Clinical Cancer Research 1, 1595-1601. Lee, S.W., Yun, M.H., Jeong, S.W., In, C.H., Kim, J.Y., Seo, M.H., Pai, C.M., Kim, S.O., 2011. Development of docetaxel-loaded intravenous formulation, Nanoxel-PM (TM) using polymer-based delivery system. Journal of Controlled Release 155, 262-271. Li, C., Cao, J., Wang, Y., Zhao, X., Deng, C., Wei, N., Yang, J., Cui, J., 2012. Accelerated blood clearance of pegylated liposomal topotecan: Influence of polyethylene glycol grafting density and animal species. Journal of Pharmaceutical Sciences 101, 3864-3876. Li, Y., Lin, T.Y., Luo, Y., Liu, Q., Xiao, W., Guo, W., Lac, D., Zhang, H., Feng, C., Wachsmann-Hogiu, S., et al., 2014. A smart and versatile theranostic nanomedicine platform based on nanoporphyrin. Nature Communications 5, 4712. Lim, J.S., Choi, J., Song, J., Chung, Y.E., Lim, S.J., Lee, S.K., Hyung, W.J., 2012. Nanoscale iodized oil emulsion: A useful tracer for pretreatment sentinel node detection using CT lymphography in a normal canine gastric model. Surgical Endoscopy 26, 2267-2274. Lin, T.Y., Zhang, H., Luo, J., Li, Y., Gao, T., Lara, P.N., Jr., de Vere White, R., Lam, K.S., Pan, C.X., 2012. Multifunctional targeting micelle nanocarriers with both imaging and therapeutic potential for bladder cancer. International Journal of Nanomedicine 7, 27932804. Lovell, J.F., Jin, C.S., Huynh, E., Jin, H., Kim, C., Rubinstein, J.L., Chan, W.C., Cao, W., Wang, L.V., Zheng, G., 2011. Porphysome nanovesicles generated by porphyrin bilayers for use as multimodal biophotonic contrast agents. Nature Materials 10, 324-332. Lu, J., Shi, M., Shoichet, M.S., 2009. Click chemistry functionalized polymeric nanoparticles target corneal epithelial cells through RGD-cell surface receptors. Bioconjugate Chemistry 20, 87-94. Lu, Z., Yeh, T.K., Wang, J., Chen, L., Lyness, G., Xin, Y., Wientjes, M.G., Bergdall, V., Couto, G., Alvarez-Berger, F., et al., 2011. Paclitaxel gelatin nanoparticles for intravesical bladder cancer therapy. Journal of Urology 185, 1478-1483. Lucas, S.R., Maranhao, R.C., Guerra, J.L., Coelho, B.M., Barboza, R., Pozzi, D.H., 2013. Pilot clinical study of carmustine associated with a lipid nanoemulsion in combination with vincristine and prednisone for the treatment of canine lymphoma. Veterinary and Comparative Oncology doi: 10.1111/vco.12033.

Page 22 of 32

601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646

MacEwen, E.G., Kurzman, I.D., Rosenthal, R.C., Smith, B.W., Manley, P.A., Roush, J.K., Howard, P.E., 1989. Therapy for osteosarcoma in dogs with intravenous injection of liposome-encapsulated muramyl tripeptide. Journal of the National Cancer Institute 81, 935-938. MacEwen, E.G., Kurzman, I.D., Vail, D.M., Dubielzig, R.R., Everlith, K., Madewell, B.R., Rodriguez, C.O., Phillips, B., Zwahlen, C.H., Obradovich, J., et al., 1999. Adjuvant therapy for melanoma in dogs: Results of randomized clinical trials using surgery, liposome-encapsulated muramyl tripeptide, and granulocyte macrophage colonystimulating factor. Clinical Cancer Research 5, 4249-4258. Marr, A.K., Kurzman, I.D., Vail, D.M., 2004. Preclinical evaluation of a liposome-encapsulated formulation of cisplatin in clinically normal dogs. American Journal of Veterinary Research 65, 1474-1478. Matsumura, Y., Maeda, H., 1986. A new concept for macromolecular therapeutics in cancer chemotherapy: Mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs. Cancer Research 46, 6387-6392. Matteucci, M.L., Anyarambhatla, G., Rosner, G., Azuma, C., Fisher, P.E., Dewhirst, M.W., Needham, D., Thrall, D.E., 2000. Hyperthermia increases accumulation of technetium99m-labeled liposomes in feline sarcomas. Clinical Cancer Research 6, 3748-3755. McCarron, P.A., Marouf, W.M., Quinn, D.J., Fay, F., Burden, R.E., Olwill, S.A., Scott, C.J., 2008. Antibody targeting of camptothecin-loaded PLGA nanoparticles to tumor cells. Bioconjugate Chemistry 19, 1561-1569. Pal, A., Khan, S., Wang, Y.F., Kamath, N., Sarkar, A.K., Ahmad, A., Sheikh, S., Ali, S., Carbonaro, D., Zhang, A., et al., 2005. Preclinical safety, pharmacokinetics and antitumor efficacy profile of liposome-entrapped SN-38 formulation. Anticancer Research 25, 331-341. Platt, S., Nduom, E., Kent, M., Freeman, C., Machaidze, R., Kaluzova, M., Wang, L., Mao, H., Hadjipanayis, C.G., 2012. Canine model of convection-enhanced delivery of cetuximabconjugated iron-oxide nanoparticles monitored with magnetic resonance imaging. Clinical Neurosurgery 59, 107-113. Poirier, V.J., Hershey, A.E., Burgess, K.E., Phillips, B., Turek, M.M., Forrest, L.J., Beaver, L., Vail, D.M., 2004. Efficacy and toxicity of paclitaxel (Taxol) for the treatment of canine malignant tumors. Journal of Veterinary Internal Medicine 18, 219-222. Poirier, V.J., Thamm, D.H., Kurzman, I.D., Jeglum, K.A., Chun, R., Obradovich, J.E., O’Brien, M., Fred, R.M., 3rd, Phillips, B.S., Vail, D.M., 2002. Liposome-encapsulated doxorubicin (Doxil) and doxorubicin in the treatment of vaccine-associated sarcoma in cats. Journal of Veterinary Internal Medicine 16, 726-731.

Page 23 of 32

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

Pradilla, G., Wang, P.P., Gabikian, P., Li, K., Magee, C.A., Walter, K.A., Brem, H., 2006. Local intracerebral administration of paclitaxel with the Paclimer® delivery system: Toxicity study in a canine model. Journal of Neuro-Oncology 76, 131-138. Rivera, P., Akerlund-Denneberg, N., Bergvall, K., Kessler, M., Rowe, A., Willmann, M., Persson, G., Kastengren Froberg, G., Westberg, S., von Euler, H., 2013. Clinical efficacy and safety of a water-soluble micellar paclitaxel (Paccal Vet) in canine mastocytomas. Journal of Small Animal Practice 54, 20-27. Rowell, J.L., McCarthy, D.O., Alvarez, C.E., 2011. Dog models of naturally occurring cancer. Trends in Molecular Medicine 17, 380-388. Schipper, M.L., Iyer, G., Koh, A.L., Cheng, Z., Ebenstein, Y., Aharoni, A., Keren, S., Bentolila, L.A., Li, J., Rao, J., et al., 2009. Particle size, surface coating, and PEGylation influence the biodistribution of quantum dots in living mice. Small 5, 126-134. Schwartz, J.A., Price, R.E., Gill-Sharp, K.L., Sang, K.L., Khorchani, J., Goodwin, B.S., Payne, J.D., 2011. Selective nanoparticle-directed ablation of the canine prostate. Lasers in Surgery and Medicine 43, 213-220. Schwartz, J.A., Shetty, A.M., Price, R.E., Stafford, R.J., Wang, J.C., Uthamanthil, R.K., Pham, K., McNichols, R.J., Coleman, C.L., Payne, J.D., 2009. Feasibility study of particleassisted laser ablation of brain tumors in orthotopic canine model. Cancer Research 69, 1659-1667. Shi, J., Zhang, J., Shen, Y., Tang, L., Zhao, J., Tu, J., Tian, Y., Feng, Y., 2013. Argininestabilized mPEG-PDLLA (50/50) polymeric micelles of docetaxel by electrostatic mechanism for tumor-targeted delivery. Drug Delivery 22, 168-181. Shin, S.J., Beech, J.R., Kelly, K.A., 2013. Targeted nanoparticles in imaging: Paving the way for personalized medicine in the battle against cancer. Integrative Biology 5, 29-42. Sorenmo, K., Samluk, M., Clifford, C., Baez, J., Barrett, J.S., Poppenga, R., Overley, B., Skorupski, K., Oberthaler, K., Van Winkle, T., et al., 2007. Clinical and pharmacokinetic characteristics of intracavitary administration of pegylated liposomal encapsulated doxorubicin in dogs with splenic hemangiosarcoma. Journal of Veterinary Internal Medicine 21, 1347-1354. Stettner, N., Brenner, O., Eilam, R., Harmelin, A., 2005. Pegylated liposomal doxorubicin as a chemotherapeutic agent for treatment of canine transmissible venereal tumor in murine models. Journal of Veterinary Medical Science 67, 1133-1139. Suzuki, T., Ichihara, M., Hyodo, K., Yamamoto, E., Ishida, T., Kiwada, H., Ishihara, H., Kikuchi, H., 2012. Accelerated blood clearance of PEGylated liposomes containing doxorubicin

Page 24 of 32

692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737

upon repeated administration to dogs. International Journal of Pharmaceutics 436, 636643. Teske, E., Rutteman, G.R., Kirpenstein, J., Hirschberger, J., 2011. A randomized controlled study into the efficacy and toxicity of pegylated liposome encapsulated doxorubicin as an adjuvant therapy in dogs with splenic haemangiosarcoma. Veterinary and Comparative Oncology 9, 283-289. Teske, E., Rutteman, G.R., vd Ingh, T.S., van Noort, R., Misdorp, W., 1998. Liposomeencapsulated muramyl tripeptide phosphatidylethanolamine (L-MTP-PE): A randomized clinical trial in dogs with mammary carcinoma. Anticancer Research 18, 1015-1019. Thamm, D.H., Vail, D.M., 1998. Preclinical evaluation of a sterically stabilized liposomeencapsulated cisplatin in clinically normal cats. American Journal of Veterinary Research 59, 286-289. U’Ren, L.W., Biller, B.J., Elmslie, R.E., Thamm, D.H., Dow, S.W., 2007. Evaluation of a novel tumor vaccine in dogs with hemangiosarcoma. Journal of Veterinary Internal Medicine 21, 113-120. Uyar, D., Kulp, B., Peterson, G., Zanotti, K., Markman, M., Belinson, J., 2004. Cardiac safety profile of prolonged (≥6 cycles) pegylated liposomal doxorubicin administration in patients with gynecologic malignancies. Gynecologic Oncology 94, 147-151. Vail, D.M., Chun, R., Thamm, D.H., Garrett, L.D., Cooley, A.J., Obradovich, J.E., 1998. Efficacy of pyridoxine to ameliorate the cutaneous toxicity associated with doxorubicin containing pegylated (Stealth) liposomes: A randomized, double-blind clinical trial using a canine model. Clinical Cancer Research 4, 1567-1571. Vail, D.M., Kravis, L.D., Cooley, A.J., Chun, R., MacEwen, E.G., 1997. Preclinical trial of doxorubicin entrapped in sterically stabilized liposomes in dogs with spontaneously arising malignant tumors. Cancer Chemotherapy and Pharmacology 39, 410-416. Vail, D.M., Kurzman, I.D., Glawe, P.C., O’Brien, M.G., Chun, R., Garrett, L.D., Obradovich, J.E., Fred, R.M., 3rd, Khanna, C., Colbern, G.T., et al., 2002. STEALTH liposomeencapsulated cisplatin (SPI-77) versus carboplatin as adjuvant therapy for spontaneously arising osteosarcoma (OSA) in the dog: A randomized multicenter clinical trial. Cancer Chemotherapy and Pharmacology 50, 131-136. Vail, D.M., MacEwen, E.G., Kurzman, I.D., Dubielzig, R.R., Helfand, S.C., Kisseberth, W.C., London, C.A., Obradovich, J.E., Madewell, B.R., Rodriguez, C.O., Jr., et al., 1995. Liposome-encapsulated muramyl tripeptide phosphatidylethanolamine adjuvant immunotherapy for splenic hemangiosarcoma in the dog: A randomized multiinstitutional clinical trial. Clinical Cancer Research 1, 1165-1170.

Page 25 of 32

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

Vail, D.M., von Euler, H., Rusk, A.W., Barber, L., Clifford, C., Elmslie, R., Fulton, L., Hirschberger, J., Klein, M., London, C., et al., 2012. A randomized trial investigating the efficacy and safety of water soluble micellar paclitaxel (Paccal Vet) for treatment of nonresectable grade 2 or 3 mast cell tumors in dogs. Journal of Veterinary Internal Medicine 26, 598-607. Venable, R.O., Worley, D.R., Gustafson, D.L., Hansen, R.J., Ehrhart, E.J., 3rd, Cai, S., Cohen, M.S., Forrest, M.L., 2012. Effects of intratumoral administration of a hyaluronancisplatin nanoconjugate to five dogs with soft tissue sarcomas. American Journal of Veterinary Research 73, 1969-1976. von Euler, H., Rivera, P., Nyman, H., Haggstrom, J., Borga, O., 2013. A dose-finding study with a novel water-soluble formulation of paclitaxel for the treatment of malignant highgrade solid tumours in dogs. Veterinary and Comparative Oncology 11, 243-255. Wang, X., Song, L., Li, N., Qiu, Z., Zhou, S., Li, C., Zhao, J., Song, H., Chen, X., 2013a. Pharmacokinetics and biodistribution study of paclitaxel liposome in Sprague-Dawley rats and Beagle dogs by liquid chromatography-tandem mass spectrometry. Drug Research 63, 603-606. Wang, Y., Liu, Y., Luehmann, H., Xia, X., Wan, D., Cutler, C., Xia, Y., 2013b. Radioluminescent gold nanocages with controlled radioactivity for real-time in vivo imaging. Nano Letters 13, 581-585. Wang, Y., Wu, K.C., Zhao, B.X., Zhao, X., Wang, X., Chen, S., Nie, S.F., Pan, W.S., Zhang, X., Zhang, Q., 2011. A novel paclitaxel microemulsion containing a reduced amount of Cremophor EL: Pharmacokinetics, biodistribution, and in vivo antitumor efficacy and safety. Journal of Biomedicine and Biotechnology 2011, 854872. Wisner, E.R., Katzberg, R.W., Link, D.P., Griffey, S.M., Drake, C.M., Vessey, A.R., Johnson, D., Haley, P.J., 1996. Indirect computed tomography lymphography using iodinated nanoparticles to detect cancerous lymph nodes in a cutaneous melanoma model. Academic Radiology 3, 40-48. Withrow, S.J., Wilkins, R.M., 2010. Cross talk from pets to people: Translational osteosarcoma treatments. ILAR Journal 51, 208-213. Working, P.K., Newman, M.S., Sullivan, T., Yarrington, J., 1999. Reduction of the cardiotoxicity of doxorubicin in rabbits and dogs by encapsulation in long-circulating, pegylated liposomes. Journal of Pharmacology and Experimental Therapeutics 289, 1128-1133. Xiao, Z., Owen, R.J., Liu, W., Tulip, J., Brown, K., Woo, T., Moore, R.B., 2010. Lipophilic photosensitizer administration via the prostate arteries for photodynamic therapy of the canine prostate. Photodiagnosis and Photodynamic Therapy 7, 106-114.

Page 26 of 32

784 785

Yu, M.K., Park, J., Jon, S., 2012. Targeting strategies for multifunctional nanoparticles in cancer imaging and therapy. Theranostics 2, 3-44.

786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804

Zahr, A.S., Davis, C.A., Pishko, M.V., 2006. Macrophage uptake of core-shell nanoparticles surface modified with poly(ethylene glycol). Langmuir 22, 8178-8185. Zhang, Z., Huey Lee, S., Feng, S.S., 2007. Folate-decorated poly(lactide-co-glycolide)-vitamin E TPGS nanoparticles for targeted drug delivery. Biomaterials 28, 1889-1899. Zhao, L., Ye, Y., Li, J., Wei, Y.M., 2011. Preparation and the in-vivo evaluation of paclitaxel liposomes for lung targeting delivery in dogs. Journal of Pharmacy and Pharmacology 63, 80-86. Zhao, Y., Wang, L., Yan, M., Ma, Y., Zang, G., She, Z., Deng, Y., 2012. Repeated injection of PEGylated solid lipid nanoparticles induces accelerated blood clearance in mice and beagles. International Journal of Nanomedicine 7, 2891-2900. Zhong, J., Mao, W., Shi, R., Jiang, P., Wang, Q., Zhu, R., Wang, T., Ma, Y., 2014. Pharmacokinetics of liposomal-encapsulated and un-encapsulated vincristine after injection of liposomal vincristine sulfate in beagle dogs. Cancer Chemotherapy and Pharmacology 73, 459-466. Figure legends

805 806

Fig. 1. Passive and active tumor targeting mechanisms of nanoparticles. Blood vessels in tumors

807

are relatively ‘leaky’ and thus allow nanoparticles to escape the blood circulation and enter

808

tumor tissues. Through surface modification with tumor targeting molecules, nanoparticles are

809

able to further specifically recognize tumor cells after extravasation. Then, through recognition

810

of the unique cancer marker, receptor-mediated endocytosis occurs. In general, active cancer

811

targeting strategy delivers greater amounts of drug and has deeper tumor penetration than

812

conventionally administered chemotherapy or uncoated nanoparticles.

813 814

Fig. 2. Active targeting of nanomicelles of canine urinary bladder cancers. The canine urinary

815

bladder cancer cell line, K9TCC-PU-Axc (kindly provided by Dr. D. Knapp from Purdue

816

University) was cultured on an eight-well chamber slide. Cells were then treated with 0.1 mg/mL

Page 27 of 32

817

PLZ4 (bladder cancer targeting peptide) coated nanomicelles (NM) loaded with DID (red

818

fluorescence dye) (PLZ4-NM-DID) or with NM-DID (non-targeting) for 20 min. Hoechst 33342

819

was used for nuclear staining (blue). Imaging was acquired using DeltaVision (GE healthcare

820

Life Science). Bar = 15m.

821 822

Page 28 of 32

823 824 825

Table 1 Nanoformulation for drug delivery, imaging, and toxicity in veterinary oncology.

Animal

Formulation/drugs

Route

Dog

Peg liposome/Doxorubicin (Doxil)

Liposome/Doxorubicin Liposome/Doxorubicin Liposome/Doxorubicin

Liposome/Muramyl tripeptide

Toxicity

References

IP

Tumor type (special application) Splenic HSA

PPES and anaphylactic reaction

IV

Non-Hodgkin’s lymphoma

Less cardiotoxicity than free doxorubicin

IV IV IV Urinary bladder submucosal injection IV

cTVT Sarcomas and carcinomas Myeloma

Grade 4 neutropenia, liver failure and renal damage

Teske et al., 2011 Sorenmo et al., 2007 Amantea et al., 1999 Working et al., 1999 Vail et al., 1998 Stettner et al., 2005 Hauck et al., 2006 Kisseberth et al., 1995 Kiyokawa et al., 1999

OSA with pulmonary metastasis Malignant melanoma Splenic HSA Appendicular OSA Mammary carcinoma Pulmonary carcinoma with metastases

Liposome/Interleukin-2 Liposome/SN-38 Liposome/Cisplatin

Inhalation IV IV

Liposome/Paclitaxel (PTX)

IV

Liposome/Vinblastine

IV

Hybrid liposome/BCNU Liposome/CPT-11 and gadoteridol

Intra-thecal Intra-tumoral

Meningeal gliomatosis Glioma (MRI)

Liposome/Cisplatin (CPI77) Liposome/Clodronate Liposome/Curcumin

IV

OSA

IV IV

Malignant histiocytosis

Peg liposome/Topotecan

IV

Liposome/SL052 Liposome-DNA complex (e.g. endostatin DNA)

IV and IA IV

Prostate cancer (PDT) HSA

Liposome/Interleukin-2 plasmid Lipid nanoemulsion/Vincristine/Pr ednisone Iodized oil emulsion

IV

Soft tissue sarcoma (Vaccine) OSA with lung metastases

IV

lymphoma

Cancerous lymph node (LN) mapping (CT lymphography)

Micelle/PTX (Paccal Vet)

Gastric submucosal injection IV

PLZ4-Micelle/PTX

IV

Nanoparticle/PTX (Crititax or CTI52010) Microsphere/PTX (Paclimer) Arginine-stabilized mPEGPDLLA polymeric micelles/Docetaxel Polymer/Docetaxel (Nanoxel-OM) Microemulsion/PTX Gelatin nanoparticles/PTX Hyaluronan conjugated cisplatin DOX-albumin

IV

Gold nanoshell

IV Intra-prostate

Mild fever post injection, and otherwise no significant toxic effects

Minimal toxicity; increased cell counts Injection site swelling and hematology changes Acute anaphylaxis-like reactions, nephrotoxicosis and substantial myelosuppression Lung nanoparticle accumulation, but no hemolysis Anorexia, weight loss, pyrexia, myelosuppression and gastrointestinal toxicity Mild lymphocytic pleocytosis in CSF, mild transient proprioceptive deficits, focal hemorrhage and focal mild perivascular non-suppurative encephalitis Acute anaphylaxis-like signs after infusion

Brief single episode of reversible hematuria; 40 mg/kg caused acute hemolysis with hematuria (oxidant effect) Accelerated blood clearance phenomenon

Grade 2/3 mast cell tumors(MCTs)

Acute urinary retention

MacEwen et al., 1989 Kleinerman et al., 1995 MacEwen et al., 1999 Vail et al., 1995 Kurzman et al., 1995 Teske et al., 1998 Khanna et al., 1997 Pal et al., 2005 Marr et al., 2004 Zhao et al., 2011 Wang et al., 2013a Kanter et al., 1994 Zhong et al., 2014 Kitamura et al., 1996 Dickinson et al., 2008, 2010 Vail et al., 2002 Hafeman et al., 2010 Helson et al., 2012 Li et al., 2012 Zhao et al., 2012 Xiao et al., 2010 U’Ren et al., 2007 Kamstock et al., 2006 Dow et al., 2005

Leucopenia

Lucas et al., 2013

Lim et al., 2012

Transient grade 3/4 neutropenia and grade 1/2 leukopenia; transient myelosuppression

High grade solid tumors Bladder cancer (mouse model)

Vail et al., 2012 Rivera et al., 2013 von Euler et al., 2013 Lin et al., 2012 Axiak et al., 2011

Intraparenchymal IV

Grade 4 neutropenia; grades 1 and 2 gastrointestinal toxicity (at high dose) Wound infection; no systemic toxicity, myelosuppression or neurotoxicity Less acute toxicity than free docetaxel

IV

Neutropenia; mild weight loss; no hypersensitivity reaction

Lee et al., 2011

Improved hypersensitivity reaction compared to free PTX

Wang et al., 2011 Lu et al., 2011 Venable et al., 2012

Systemic histamine-like reaction after injection at high dose; no toxicity at low dose

Kratz et al., 2007

IV Intra-vesical Intra-tumoral

Bladder cancer Soft tissue sarcomas

IV Orthotopic cTVT in brain Normal prostate (NIR imaging and PTT)

Pradilla et al., 2006 Shi et al., 2013

Schwartz et al., 2009 Schwartz et al., 2011

Page 29 of 32

Cat

Rabbit Pig

826 827 828 829 830

Gold nanoparticle

Intra-tumoral

Cetuximab conjugated iron oxide nanoparticles Doxil

Intra-cranial

Thyroid carcinoma and OSA (CT scan) MRI

Chanda et al., 2014

IV

Vaccine-associated sarcoma

Liposome/Doxorubicin + radiotherapy Liposome/MTP

IV

Soft tissue sarcomas

Kleiter et al., 2010

IV

Mammary adenocarcinoma

Fox et al., 1995

Liposome/Cisplatin Liposome/Cis-bisneodecanoato-trans-R,R-1,2diaminocyclohexane platinum (II) Liposome/Meta(tetrahydroxyphenyl)chlorin e Magnetic nanoparticles/feline granulocyte macrophagecolony stimulating factor plasmid or interleukin-2 or interferon-γ Liposome/Technetium-99m Doxil Lymphotropic iodinated nanoparticle

IV IV

None

Platt et al., 2012

Delayed nephrotoxicosis; localized hyperpigmentation and alopecia (chin); mild transient gastrointestinal signs; sudden explained death

Vail et al., 1997 Poirier et al., 2002

Increased cholesterol levels 2 days after injection; pyrexia Transient pyrexia; lethargy; vomiting; inappetence; acute infusion reaction; thrombocytopenia; cumulative myelosuppression; liver/kidney toxicity

Thamm and Vail, 1998 Fox et al., 1999

IV

Cutaneous squamous cell carcinoma (PDT)

Mild local toxicity, such as erythema and edema

Buchholz et al., 2007

Intra-tumoral

Fibrosarcoma

None

Jahnke et al., 2007 Huttinger et al., 2008

IV IV Peri-tumoral

Sarcoma (planar scintigraphy/PET) Progressive cardiomyopathy Cutaneous malignant melanoma (cancerous lymph node mapping)

Matteucci et al., 2000 Working et al., 1999 Wisner et al., 1996

IV, intravenous injection; IP, intraperitoneal injection; IA, intra-arterial injection; BCNU, 1,3-bis(2-chloroethyl)-1-nitrosourea; HSA, hemangiosarcoma; cTVT, canine transmissible venereal tumor; OSA, osteosarcoma; BAL, bronchoalveolar lavage; CSF, cerebrospinal fluid; PPES, palmar-plantar erythrodysesthesia; PDT, Photodynamic therapy; PTT, Photothermal therapy; MRI, Magnetic resonance imaging; CT, computed tomography; NIR, Near infrared red.

Page 30 of 32

831 832 833

Table 2 Selective clinical trials on nanoformulations in veterinary medicine.

Animal Dog/cat

Nanoformulation Cisplatin hyaluronan nanoparticles

Dog

Gold nanorod mediated photothermal therapy Radioactive gold nanoparticles Temozolomide loaded polycaprolactone magnetite nanoparticles Doxorubicin loaded nanoparticle Liposomal curcumin Liposomal clodronate Paccal Vet (micellar paclitaxel)

Route Intra-tumoral and peri-tumoral IV

Tumor type Oral squamous cell carcinoma and malignant melanoma Solid tumor

Organization University of Missouri

IV IV

Primary prostate tumor Glioma

IV IV IV IV

Appendicular osteosarcoma Lung cancer Soft-tissue sarcoma Histiocytic sarcoma (phase II)

University of Missouri University of Illinois and University of Chicago University of Illinois University of California-Davis Colorado State University University of Tennessee at Knoxville

Ohio State University

834 835

Page 31 of 32

836

Page 32 of 32

Nanomedicine in veterinary oncology.

Nanomedicine is an interdisciplinary field that combines medicine, engineering, chemistry, biology and material sciences to improve disease management...
802KB Sizes 5 Downloads 20 Views