Targeted Tubulysin B Hydrazide Conjugate for the Treatment of

May 31, 2018 - The targeted delivery of chemotherapeutic agents to receptors that are overexpressed on cancer cells has become an attractive strategy ...
0 downloads 0 Views 909KB Size
Subscriber access provided by Kaohsiung Medical University

Article

A targeted tubulysin B hydrazide conjugate for the treatment of LHRH receptor-positive cancers. jyoti roy, Miranda Kaake, Madduri Srinivasarao, and Philip S. Low Bioconjugate Chem., Just Accepted Manuscript • DOI: 10.1021/acs.bioconjchem.8b00164 • Publication Date (Web): 31 May 2018 Downloaded from http://pubs.acs.org on June 1, 2018

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 18 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

Bioconjugate Chemistry

A targeted tubulysin B hydrazide conjugate for the treatment of LHRH receptor-positive cancers.

1 2 3

Jyoti Roy1,2, Miranda Kaake2, Madduri Srinivasarao1,2, and Philip S Low1,2,*

4

1

Purdue Institute for Drug Discovery, Purdue University, West Lafayette IN 47907 USA

5

2

Department of Chemistry, Purdue University, West Lafayette IN 47907 USA

6

*Author to whom all correspondence should be addressed:

7

Philip S Low

8

Email: [email protected]

9

Phone: (765)494-5283

10

Fax: (765)496-2677

11

Department of Chemistry

12

Purdue University

13

720 Clinic Dr.

14

West Lafayette IN 47907

15

The authors declare no potential conflicts of interest.

16 17 18 19 20 21 22 23 24 25 1 ACS Paragon Plus Environment

Bioconjugate Chemistry 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

26

Abstract:

27

The targeted delivery of chemotherapeutic agents to receptors that are over-expressed on cancer

28

cells has become an attractive strategy to concentrate drugs in cancer cells while avoiding uptake

29

by healthy cells. Luteinizing hormone-releasing hormone receptor (LHRH-R) has attracted

30

considerable interest for this application, since LHRH-R is upregulated in ~86% of prostate

31

cancers, ~80% of endometrial cancers, ~80% of ovarian cancers, and ~50% of breast cancers, but

32

virtually absent from normal tissues. Although LHRH and related peptides have been used to

33

deliver cytotoxic drugs to LHRH-R over-expressing cancer cells, they have suffered from off-

34

target delivery of the therapeutic agents to the liver and kidneys. To reduce such unwanted

35

uptake by peptide scavenger receptors in the liver and kidneys, we have explored the use of a

36

nonpeptidic LHRH-R antagonist (NBI42902) to construct an LHRH-R targeted tubulysin

37

conjugate (BOEPL-L3-TubBH). In vitro studies with BOEPL-L3-TubBH demonstrate that the

38

conjugate can kill LHRH-R expressing triple-negative breast cancer cells (MDA-MB-231 cells)

39

with low nanomolar IC50. Related studies on tumor-bearing mice further reveal that the same

40

conjugate can eradicate MDA-MB-231 solid tumors without any measurable side-effects,

41

yielding mice that gain weight during therapy and show no evidence of tumor recurrence for at

42

least 5 weeks after termination of treatment. That these complete responses are LHRH-R targeted

43

was then established by showing that identical treatment of receptor-negative (SKOV3) tumors

44

yields no anti-tumor response. Overall, these data provide a proof-of-concept that LHRH-R

45

specific targeting of an extremely toxic drug like tubulysin B can treat LHRH-R positive tumors

46

without causing significant toxicity to healthy cells.

47 48 49 50 51 52 53 54 55 56

2 ACS Paragon Plus Environment

Page 2 of 18

Page 3 of 18 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

Bioconjugate Chemistry

57 58

Introduction:

59

Because nontargeted chemotherapeutic agents exhibit little cell type specificity, they distribute

60

indiscriminately into most cells of the body, often causing toxicity to cancer cells and healthy

61

cells alike.1,2 One strategy to limit adverse side effects of nontargeted drugs is to tether the

62

cytotoxic agent to a targeting ligand that can carry the therapeutic cargo selectively to malignant

63

cells, thereby avoiding most uptake by healthy cells. To achieve such tumor-specific targeting, it

64

is usually necessary to identify a receptor that is over-expressed on the cancer cells, but largely

65

absent from normal cells. Construction of a ligand that can deliver an attached drug directly to

66

this receptor should then enable selective accumulation of the cytotoxic drug within the

67

malignant tissue.

68

One receptor known to be over-expressed in many cancers is the G-protein coupled receptor,

69

luteinizing hormone-releasing hormone receptor (also known as gonadotropin-releasing hormone

70

receptor).3,4 Luteinizing hormone-releasing hormone receptor (LHRH-R) is upregulated on many

71

cancers of the breast (50%),5-8 prostate (86%),8-12 endometrium (80%),12,14-17 and ovary (80%)12-

72

15,17

73

contrast, the same receptor is essentially absent from healthy tissues except for the pituitary

74

gland and reproductive organs, where it naturally functions to induce the synthesis and secretion

75

of such gonadotropins as luteinizing hormone (LH) and follicle-stimulating hormone (FSH).

76

Because these gonadotropins, in turn, promote the production of steroid sex hormones, it is not

77

surprising that estrogen- and androgen-dependent cancers often depend on LHRH-R signaling

78

for proliferation and survival.3,4

79

LHRH peptide and its analogs have been extensively used as targeting ligands to deliver a range

80

of cytotoxic drugs to tumors expressing the LHRH receptor. Several such LHRH conjugates are

81

in the early stages of development, whereas others have already advanced to human clinical

82

trials. AEZS-108 (AN-152), for example, is an LHRH-targeted doxorubicin conjugate that is

83

currently in phase III clinical trials for the treatment of endometrial cancer and in phase II

84

clinical trials for the therapy of ovarian cancer.6,14,21 EP-100, in contrast, consists of the LHRH

85

hormone linked to a toxic membrane-disrupting peptide that is currently in phase II clinical trials

86

for the treatment of advanced LHRH-R positive tumors.22 LHRH-targeted drugs undergoing

and also on some non-hormone dependent cancers of the pancreas,18,19 skin,20 and brain.21 In

3 ACS Paragon Plus Environment

Bioconjugate Chemistry 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

87

preclinical development include the anti-androgen, CBDIV17, encapsulated in LHRH-

88

derivatized micelles,23 and multiple LHRH peptides/analogs linked to the cytotoxic drugs

89

cisplatin,24 disorazol,25 ERK inhibitors, PI3K/ERK inhibitors5, or gemcitabine26. Finally,

90

researchers have also developed and tested LHRH-targeted paclitaxel-loaded microbubbles for

91

ultrasound-stimulated drug release within a tumor mass.27

92

Although the above LHRH-targeted therapeutic conjugates have shown promise in treating

93

LHRH-R expressing tumors, such peptide-derived drugs often display limitations that can

94

adversely impact their tumor specificities. First, because the natural targeting ligand is rapidly

95

metabolized in vivo, its premature degradation before arrival in the tumor mass can compromise

96

tumor specificity. Second, due to elevated expression of peptide scavenger receptors in the liver,

97

kidneys, and other organs, capture and retention of the peptide-targeted conjugates can often

98

occur in healthy organs, causing organ damage even in the absence of a targeted receptor.28,29 In

99

order to avoid these unwanted side effects, we have undertaken to design a non-peptidic,

100

proteolytically stable, small molecule LHRH-R targeting ligand that can deliver an attached

101

therapeutic payload selectively to LHRH-R over-expressing cancers without depositing

102

measurable drug in the liver or kidneys, etc. We demonstrate below that conjugation of this

103

ligand to the microtubule inhibitor, tubulysin B hydrazide, creates an LHRH-R targeted drug that

104

can eradicate LHRH-R expressing cancer cells both in vitro and in vivo.

105

Results:

106

Design and Synthesis of LHRH-R conjugates. To develop LHRH-R targeted tubulysin conjugate

107

we searched the literature for a high-affinity LHRH-R antagonist that would also inhibit LHRH

108

signaling. Although the LHRH antagonist, NBI42902, met these criteria and exhibited a high

109

affinity (Kd= 0.19 nM; Ki= 0.56 nM) for LHRH-R,30 the non-peptidic antagonist could not be

110

used directly for drug targeting because it lacked a site for facile conjugation of a therapeutic

111

cargo (Fig. 1).

112

NBI42902 that could be readily modified without loss of antagonist activity.30 This search

113

revealed that the ether functionality on the fluorinated aromatic ring (See Supplementary Fig. S1)

114

could be substituted without significant impact on LHRH-R binding affinity,30 and for ease of

115

derivatization, we elected to replace this aliphatic ether with an aliphatic carboxylic acid (see

116

BOEPL, Fig. 1). The modified ligand, which we named breast, ovarian, endometrial, and

To introduce such a ligation site into NBI42902, we looked for sites on

4 ACS Paragon Plus Environment

Page 4 of 18

Page 5 of 18 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

Bioconjugate Chemistry

117

prostate cancer ligand (BOEPL), was used for the synthesis of all conjugates reported in this

118

article. Finally, to enhance the solubility of these conjugates and to reduce their nonspecific

119

uptake by receptor negative cells, the ligand was conjugated to its payload via a hydrophilic

120

linker, as shown in Fig. 1.

121

122 123

Figure 1. Chemical structures. Structure of LHRH-R targeting ligand NBI42902 and its

124

modified version BOEPL, free tubulysin B hydrazide (TubBH), LHRH-R targeting ligand

125

conjugated to peptidoglycan linker (BOEPL-L3), LHRH-R targeted tubulysing B hydrazide

126

conjugate (BOEPL-L3-TubBH), LHRH-R targeting ligand conjugated to PEG linker (BOEPL-

127

L2), and LHRH-R targeted rhodamine conjugate (BOEPL-L2-Rhodamine).

128

For the studies below, two different conjugates had to be synthesized: i) a BOEPL-rhodamine

129

conjugate for use in evaluating cell binding and internalization, and ii) a BOEPL-tubulysin B

130

hydrazide conjugate for analysis of tumor-specific cytotoxicity. The rhodamine conjugate was

131

prepared by attaching the dye to the BOEPL ligand via a non-cleavable hydrophilic linker to

132

assure that the location of the conjugate could be accurately tracked by following the

133

rhodamine’s fluorescence. In contrast, the tubulysin B hydrazide had to be synthesized by

134

tethering the drug to the ligand via a self-immolative disulfide linker (Fig. 1) to allow for facile

135

release of the unmodified microtubule inhibitor upon entry of the conjugate into a reducing

136

endosome within the target cell. Moreover, because the tubulysin conjugate of BOEPL was 5 ACS Paragon Plus Environment

Bioconjugate Chemistry 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

137

much more hydrophobic than the rhodamine conjugate of the same ligand, the hydrophilic

138

linkers tethering BOEPL to its two payloads had to be independently optimized for each payload.

139

Binding and Internalization of rhodamine dye conjugate: Binding and uptake of the LHRH-R

140

targeted rhodamine conjugate was evaluated by incubating MDA-MB231 or SKOV3 cancer cells

141

for 1h with increasing concentrations of BOEPL-L2-rhodamine either in the presence or absence

142

of excess competing for BOEPL-L2. After washing away excess conjugate, cells were

143

immediately examined by confocal microscopy to determine the location of any cell-associated

144

fluorescence. As shown in Fig. 2, rhodamine conjugate was internalized primarily within the

145

intracellular compartments of the MDA-MB231 cells, with little fluorescence remaining on the

146

cell surfaces. Total absence of intracellular and cell surface fluorescence in samples treated with

147

a 100-fold excess of non-fluorescent BOEPL-L2 demonstrated that this cell-associated

148

fluorescence was LHRH receptor mediated. This conclusion was further substantiated by that

149

absence of any BOEPL-L2-rhodamine binding or internalization in LHRH receptor negative

150

SKOV3 cells (Fig. 2). Importantly, the nearly quantitative intracellular localization of the

151

targeted rhodamine conjugate following only 1 hour of incubation suggests that receptor-

152

mediated endocytosis of BOEPL-L2-rhodamine is rapid in MDA-MB231 cells. Such rapid

153

internalization can be beneficial to drug delivery, since it entraps the payload inside the target

154

cell.

155 156

Figure 2. Confocal Imaging. In vitro binding of BOEPL-L2-Rhodamine conjugate to MDA-MB-

157

231 (LHRH-R positive) and SKVO3 (LHRH-R negative) cells either in the presence or absence

158

of excess of BOEPL-L2 (n=2). 6 ACS Paragon Plus Environment

Page 6 of 18

Page 7 of 18 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

Bioconjugate Chemistry

159

In vitro cytotoxicity of tubulysin B hydrazide conjugate: To explore the killing efficiency of the

160

BOEPL-L3-tubulysin conjugate, MDA-MB231 and SKOV3 cancer cells were incubated with

161

various concentrations of BOEPL-L3-TubBH, either in the presence or absence of 100-fold

162

excess BOEPL-L3 to compete for conjugate binding. As shown in Fig. 3, both MDA-MB231

163

and SKOV3 cells were very sensitive to free tubulysin B hydrazide, yielding IC50 values of 67

164

pM and 5 pM, respectively. In contrast, only the LHRH-R positive MDA-MB231 cells were

165

killed by receptor-targeted tubulysin B hydrazide (IC50 ~ 26 nM) and this cytotoxicity could be

166

blocked by addition of 100-fold excess BOEPL-L3. As anticipated, receptor negative SKOV3

167

cells were largely insensitive to BOEPL-L3-TubBH, suggesting that uptake of the targeted

168

cytotoxic conjugate requires binding and endocytosis by the LHRH receptor. The fact that

169

SKOV3 cells were readily killed by free tubulysin hydrazide but not by its targeted conjugate

170

further reveals that the hydrophilic conjugate is too polar and/or large to diffuse passively into

171

receptor-negative cells. This latter property is critical to the prevention of off-target toxicity

172

since the self-immolative linker is designed to release free tubulysin hydrazide immediately upon

173

entry into a reducing environment such as the interior of virtually all mammalian cells.

174 175

Figure 3. Potency of TubBH and BOEPL-L3-TubBH. In vitro cytotoxicity assay showing dose

176

response activity of TubBH (black triangle) and LHRH-R targeted tubulysin B hydrazide

177

conjugate (BOEPL-L3-TubBH) either in the presence (grey circle) or absence (black square) of

7 ACS Paragon Plus Environment

Bioconjugate Chemistry 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

178

excess of BOEPL-L3 in LHRH-R positive MDA-MB-231 (A) and LHRH-R negative SKOV3

179

(B) cells (n=3).

180

In vivo efficacy of tubulysin B hydrazide conjugate: To investigate the efficacy of BOEPL-L3-

181

TubBH conjugate in nu/nu athymic nude mice bearing LHRH-R positive tumors (MDA-

182

MB231), mice were treated 3x/week for 3 weeks with either saline control, targeted BOEPL-L3-

183

TubBH, or targeted BOEPL-L3-TubBH in the presence of 100-fold excess of free BOEPL-L3.

184

As shown in Fig. 4, MD-MB231 tumor bearing mice that received targeted tubulysin B

185

hydrazide showed complete elimination of their tumors, whereas mice treated with saline

186

displayed rapid tumor growth. Similarly, mice co-injected with excess competing ligand

187

exhibited no suppression of tumor growth compared to the control group (Fig. 4A). These

188

strongly argue that the efficacy of the LHRH receptor-targeted tubulysin B hydrazide is receptor

189

mediated. However, to unequivocally establish that the anti-tumor efficacy of the targeted

190

conjugate (BOEPL-L3-TubBH) is LHRH-R mediated, mice bearing LHRH-R negative tumors

191

(SKOV3, ovarian cancer) were divided into analogous treatment groups and monitored similarly.

192

As expected, no reduction in tumor volume was observed in either the targeted or control groups,

193

despite the fact that SKOV3 tumors are very tubulysin B hydrazide sensitive (Fig. 4C); i.e.

194

demonstrating that the toxicity seen in MDA-MB-231 tumors was not due to nonspecific

195

systemic release of free tubulysin B hydrazide. Finally, to explore the effect of the targeted

196

tubulysin B hydrazide conjugate on systemic toxicity, body weights of mice were monitored over

197

the course of the study. As seen in Fig. 4B & 4D, none of the mice bearing either MDA-MB-231

198

or SKVO3 tumors experienced significant weight loss, suggesting that the targeted conjugate is

199

not toxic to either receptor positive or negative cancers.

200

After completion of the treatment, mice in the MDA-MB231 targeted group were monitored with

201

no further therapy for an additional five weeks. During this time the tumor volume and weight

202

were monitored three time a week (Fig. 5). None of the mice showed any indication of tumor

203

growth or weight loss. These data suggest that the LHRH-R targeted tubulysin B hydrazide

204

conjugate (BOEPL-L3-TubBH) constitutes a promising candidate for treatment of cancers that

205

over-express LHRH receptors.

8 ACS Paragon Plus Environment

Page 8 of 18

Page 9 of 18 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

Bioconjugate Chemistry

206 207

Figure 4. Therapeutic efficacy of BOEPL-L3-TubBH. LHRH-R positive (MDA-MB-231) and

208

negative (SKOV3) cancer cells were s.c implanted in to nu/nu athymic mice and treatment was

209

initiated once the tumor volume reached ~100 mm3. Mice were randomized into several groups

210

with 5 mice in each group. Mice in control group received saline, whereas mice in the

211

competition and treatment group received 2 µmol/kg of BOEPL-L3-TubBH either in the

212

presence or absence of 100-fold excess of BOEPL-L3 respectively for 3 times per week for 3

213

weeks. All the test agents were administered intravenously through tail vein. A and C represent

214

tumor volume (mm3), and B & D represent animal body weight (g).

215 216

Figure 5. Therapeutic efficacy of BOEPL-L3-TubBH against subcutaneous LHRH-R positive

217

MDA-MB231 tumor growth. Mice were treated with the LHRH-R targeted tubulysin B

218

hydrazide conjugate (BOEPL-L3-TubBH) for thrice a week for 5 weeks (n=5). After completion

219

of the study, tumor volume (A; mm3) and animal body weight (B; g) were continuously

9 ACS Paragon Plus Environment

Bioconjugate Chemistry 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

Page 10 of 18

220

monitored for 75 days post tumor implantation. The dotted vertical line specifies the last day of

221

dosing.

222

Discussion:

223

All LHRH receptor targeted drug conjugates described to date employ an LHRH peptide or one

224

of its analogs as the targeting ligand. However, because such peptides are rapidly digested or

225

captured by scavenger receptors in the liver and kidneys, concerns can logically be raised that

226

cytotoxic conjugates of an LHRH peptide might cause damage to one or both organs28-29. Indeed,

227

when normal rats were injected intravenously with

228

the injected dose was observed two hours later in the kidneys (>5% ID/g), liver (>2% ID/g),

229

stomach (>6% ID/g), and intestines (>4 % ID/g), suggesting significant uptake of LHRH is

230

indeed observed in healthy tissues.31, 32 Although a small fraction of this retention could be due

231

to slow clearance of the decapeptide from the liver and kidneys, when linked to a cytotoxic drug

232

for cancer therapy, the unwanted accumulation of drug in these normal organs could cause

233

unnecessary toxicity. This concern is especially worrisome when tubulysin is used as the

234

warhead, since tubulysins in their nontargeted forms have proven to be undosable in vivo due to

235

severe toxicities to malignant and healthy cells alike.33 Our strategy for avoiding such off-target

236

toxicities was to conjugate tubulysin to a modified version of the nonpeptidic LHRH-R

237

antagonist, NBI42902 (K(i) ~0.56 nM).30 However, due to lack of an appropriate functional

238

group, derivatization of NBI42902 proved to be difficult, but could be overcome by replacing the

239

ether group on NBI42902 with a carboxyl group. Because the ether formed part of the molecule

240

that does not interact with the receptor, the selectivity and specificity of the resulting conjugate

241

for LHRH-R was not remarkably different from the parent antagonist. Moreover, our ability to

242

target tubulysin B hydrazide to LHRHR positive tumors without causing weight loss argues

243

strongly that the conjugate was primarily concentrated in the tumor masses. In fact, when we

244

prepared and tested a conjugate of the same targeting ligand linked to a

245

conjugate revealed little or no uptake in the liver or GI tract and only temporary uptake in the

246

kidneys (unpublished data).

247

While the absence of overt toxicity constitutes an essential property of a viable cancer drug

248

candidate, lack of toxicity alone is insufficient to justify development of a therapeutic lead. In the

249

paragraphs above, we have documented that the tubulysin B conjugate of the NBI42902

99m

Tc-labeled LHRH, a high percentage of

10 ACS Paragon Plus Environment

99m

Tc chelate, the

Page 11 of 18 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

Bioconjugate Chemistry

250

derivative (BOEPL-L3-TubBH) can completely eradicate LHRH-R positive human tumor

251

xenografts in athymic mice. Thus, whereas the other reported LHRH-R targeted therapies have

252

only succeeded in slowing tumor growth,6,21-25 BOEPL-L3-Tubulysin was found to yield

253

complete responses that persisted for at least 5 weeks following termination of treatment (i.e. the

254

length of the monitoring period). Taken together with cytotoxicity data on i) LHRH receptor

255

negative tumors (SKOV3), and ii) LRHR positive tumors that were blocked with excess ligand

256

(BOEPL-L3), our results demonstrate that the therapeutic efficacy of BOEPL-L3-TubBH is

257

indeed receptor-mediated. While the tumor specificity of such a targeted drug should almost

258

always improve the toxicity profile of the drug, it should also be remembered that the receptor

259

targeting strategy will simultaneously impose a limitation on the amount of drug that can be

260

delivered, since this amount will invariably be determined by the number of receptors per cancer

261

cell and the rate of receptor recycling into intracellular compartments and back to the cell

262

surface.34,35

263

Because virtually all naturally occurring tumors are heterogeneous, monotherapies seldom yield

264

sustained responses in humans, prompting oncologists to search for synergistic combination

265

therapies to treat most cancers. Fortunately, multiple established treatment options already exist

266

for cancers that overexpress LHRH-R, yielding many FDA-approved drugs that might be mixed

267

with BOEPL-L3-tubulysin and examined for synergy in the treatment of prostate8-12

268

endometrial,12,14-17 ovarian,2,15-17 and breast5-8 cancers. Thus, because LHRH stimulation of

269

LHRH receptor is required for sex steroids production and since these same hormones can drive

270

progression of breast, ovarian, endometrial and prostate cancers, especially during early stages of

271

the respective diseases, conjugation of an LHRHR antagonist (i.e. NBI42902) to a highly

272

cytotoxic drug might not only inhibit production of tumor-sustaining sex steroids, but also kill

273

malignant cells that over-express the LHRH receptor. It will obviously be important in future

274

studies to explore the impact of BOEPL-L3-TubBH therapy on androgen and estrogen

275

production and to determine whether the BOEPL-L3-TubH conjugate might suppress

276

reproductive tumor growth by multiple mechanisms of action. It will also be important to

277

evaluate the pharmacokinetics and pharmacodynamics of the conjugate and any metabolites to

278

determine whether it might qualify for further development.

279

Experimental Procedure:

11 ACS Paragon Plus Environment

Bioconjugate Chemistry 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

Page 12 of 18

280

Materials: H-Cys(Trt)-2-Cl-Trt resin and protected amino acids were purchased from Chem-

281

Impex

282

hexafluorophosphate

283

dimethylformamaide (DMF), piperidine, isopropryl alcohol (IPA), dichloromethane (DCM),

284

dmithelysulfoxide (DMSO) and most other chemicals were purchased from Sigma Aldrich.

285

Tubulysin B hydrazide and its activated derivative was a kind gift from Endocyte Inc. (West

286

Lafayette, IN). RPMI media and fetal bovine serum (FBS) were purchased from GIBCO (Grand

287

Island, NY). Glutamine, penicillin-streptomycin and trypsin were procured from BD Biosciences

288

(San Jose, CA).

289

Conjugate Synthesis:

290

Synthesis of BOEPL-L2: The LHRH-R antagonist (BOEPL) was synthesized per the previously

291

published procedure30 except a carboxylic acid derivative was used in the last step instead of the

292

ether shown Supplemental Fig. 1. This modification was introduced to enable its subsequent

293

coupling to a linker. The desired linker was prepared by standard solid phase peptide synthesis as

294

shown in supplementary Fig. S1, after which BOEPL was coupled via its carboxyl to the linker.

295

The product, BOEPL-L2 (Fig. 1) was then cleaved from the resin using a hydrolysis solution

296

comprised of TFA:water:triisopropylsilane:ethanedithiol (95%: 2.5%: 2.5%: 2.5%) and the crude

297

BOEPL-L2 conjugate was purified by reverse phase HPLC [A=2 Mm ammonium acetate buffer

298

(pH 5.0), B= acetonitrile, solvent gradient 0% B to 80% B in 35 min] to afford the desired

299

product at 70% yield. LRMS-LC/MS (m/z): [M+H]+ calcd for C47H61F2N5O12S, 958.08; found

300

959.

301

Synthesis of BOEPL-L2-Rhodamine: To synthesize the rhodamine conjugate of BOEPL-L2,

302

purified BOEPL-L2 and rhodamine maleimide (1 eq) were dissolved in anhydrous DMSO

303

containing DIPEA (2 eq). The reaction mixture was stirred under argon atmosphere at room

304

temperature (See supplementary Fig. S1) during which the reaction’s progress was monitored by

305

analytical LRMS-LCMS. Following completion of the reaction (~1h) crude product was purified

306

by preparative RP-HPLC [A= 2mM ammonium acetate buffer (pH 7.0), B=acetonitrile, solvent

307

gradient 0% B to 50% B in 35 min] to yield 90% of the desired product. LRMS-LC/MS (m/z):

308

[M+H]+ calcd for, C75H84F2N8O17S, 1439.59; found 1440.

Intl.

(Chicago,

IL).

2-(1H-7-azabenzotriazole-1-yl)-1,1,3,3-tetramethyl

methanaminium

(HATU),

diisopropylethylamine

12 ACS Paragon Plus Environment

uronium

(DIPEA),

N,N’

Page 13 of 18 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

Bioconjugate Chemistry

309

Synthesis of BOEPL-L3: BOEPL-L3 was prepared by the standard solid phase peptide synthesis

310

described in supplementary Fig. S2, after which the final product was cleaved from the resin as

311

described above. Crude BOEPL-L3 was purified by RP-HPLC [A=2 Mm ammonium acetate

312

buffer (pH 5.0), B= acetonitrile, solvent gradient 0% B to 80% B in 35 min] to yield 80% of the

313

desired product. LRMS-LC/MS (m/z): [M+H]+ calcd for C67H94F2N10O23S, 1477.59; found 1478.

314

Synthesis of BOEPL-L3-TubBH: Tubulysin B hydrazide was synthesized as described below.

315

Briefly, BOEPL-L3 was dissolved in argon purged HPLC grade water and adjusted to pH7.0

316

using a NaCO3-saturated solution of the same water. To this reaction mixture was added

317

disulfide activated tubulysin B hydrazide (1 eq) in THF and the reaction mixture was allowed to

318

stir at room temperature under argon atmosphere. The progress of the reaction was monitored

319

using analytical LRMS-LCMS, and after its completion (~30 min.) the crude product was

320

purified by preparative RP-HPLC [A=2 Mm ammonium acetate buffer (pH 7.0), B= acetonitrile,

321

solvent gradient 0% B to 80% B in 35 min] to yield 90% of the desired product. LRMS-LC/MS

322

(m/z): [M+H]+ calcd for C112H161F2N17O34S3, 2423.78; found 2424.

323

Cell culture and confocal microscopy: LHRH-R positive breast cancer cells (MDA-MB231) and

324

LHRH-R negative ovarian cancer cells (SKVO3) were cultured in RPMI 1640 medium

325

supplemented with 10% fetal bovine serum and 1% penicillin–streptomycin at 37 ˚C in a 95%

326

humidified air and 5% CO2 atmosphere. MDA-MB231 and SKVO3 cells were seeded into

327

chambered coverglass plates and allowed to grow to confluence over 48 h. Spent medium was

328

replaced with 0.5 mL of fresh medium and incubated with 100 nM of the LHRH-R targeted

329

rhodamine dye (BOEPL-L2 -rhodamine) either in the presence or absence of 100-fold excess of

330

BOEPL-L2. After incubation for 1h, the cells were washed 3x in incubation solution and then

331

replaced with 0.5 ml of fresh culture medium. Images were acquired using Olympus confocal

332

microscopy.

333

The cell culture of MDA-MB231 and SKOV3 cell lines used in this study was initiated by

334

thawing frozen vials (2016) from a master stock saved from the original cell lines previously

335

purchased from ATCC. All experiments were performed within two or three passages following

336

thawing of the cell lines. No mycoplasma test was performed on both the cell lines.

337

In vitro determination of cell viability. MDA-MB231 and SKVO3 cells were seeded at a

338

concentration of 100,000 cells per well on a 24 well plate and allowed to grow in monolayers. 13 ACS Paragon Plus Environment

Bioconjugate Chemistry 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

339

Spent medium was removed and cells were incubated in various concentrations of BOEPL-L3-

340

TubBH either in the presence or absence of 100-fold excess of BOEPL-L3 in fetal bovine serum

341

free medium. After incubating for 2 h at 37 ˚C, cells were rinsed three times with fresh medium

342

and then incubated in 0.5 ml fresh medium for an additional 66 h at 37 ˚C. Spent medium was

343

then replaced with 0.5 ml of fresh medium containing 3H-thymidine and incubated for additional

344

4 h. Cells were again washed 3x with fresh medium and then incubated with 0.5ml of 2.5 %

345

trichloroacetic acid for 10 min at room temperature. After removing the trichoroacetic acid, cells

346

were dissolved in 0.25 N NaOH. Cell viability was determined by counting the incorporation of

347

3

348

IC50 value of the BOEPL-L3-TubBH was derived from a plot of the percent of 3H-thymidine

349

incorporation versus log concentration using Graph Pad Prism 4.

350

Animal husbandry and therapy: 4-5-week-old Athymic nu/nu female mice were purchased from

351

Harlan Laboratories. Mice were housed in a sterile environment on a standard 12-hour light–dark

352

cycle and maintained on normal rodent chow. All animal procedures were approved by the

353

Purdue Animal Care and Use Committee in accordance with National Institutes of Health

354

guidelines. 5-6 weeks old female nu/nu athymic nude mice were subcutaneously injected with

355

5.0 × 106 breast cancer cells (MDA-MB231) or ovarian cancer cells SKVO3 into their shoulders.

356

Tumors were measured in two perpendicular directions 3 times per week with Vernier calipers,

357

and their volumes were calculated as 0.5 x L x W2, where L is the longest axis (in millimeters),

358

and W is the axis perpendicular to L (in millimeters). Treatment was initiated once the tumor

359

volume reached ∼100 mm3. Dosing solutions were prepared in sterile saline and injected

360

intravenously. Each mouse received 2µmol/kg of BOEPL-L3-TubBH either in the presence or

361

absence of 100-fold excess of BOEPL-L3 or saline. Mice were administered with the test agents

362

3x per week for 3 weeks and weighed as a measure of drug toxicity at each dosing.

363

Supporting Information:

364

Synthetic schemes and characterization of BOEPL-L2-Rhodamine, BOEPL-L3, and BOEPL-L3-

365

TubBH.

366

Acknowledgment: Authors would like to thank Endocyte Inc. (West Lafayette, IN) for funding

367

this research work.

H-thymidine in cells using a scintillation counter (Packard, Packard Instrument Company). The

14 ACS Paragon Plus Environment

Page 14 of 18

Page 15 of 18 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

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

Bioconjugate Chemistry

References: 1. Azim, H. A. Jr., de Azambuja E., Colozza, M., Bines, J., Piccart, M. J. Long-term toxic effects of adjuvant chemotherapy in breast cancer. Ann Oncol. 22, 1939-1847 (2011). 2. Allen, T. M. Ligand targeted therapeutics in anticancer therapy. Nat Rev Cancer. 2, 750763 (2002). 3. Chillik, C., Acosta, A. The role of LHRH agonists and antagonists. Repro Biomed online. 2, 120-128 (2001). 4. Tolkach, Y., Joniau, S., Van Poppel, H. Luteinizing hormone-releasing hormone (LHRH) receptor agonists vs antagonists: a matter of the receptors? BJU Int. 111, 1021-1030 (2013). 5. Kwok, C. K., Treeck, O., Buchholz, S., Ortmann, O., Engel, J. B. Receptors for luteinizing hormone-releasing hormone (GnRH) as therapeutic targets in triple negative breast cancers (TNBC). Targ Oncol. 10, 365-373 (2015). 6. Seitz, S. et al. Triple Negative breast cancers express receptors for LHRH and are potential therapeutic targets for cytotoxic LHRH-analogs, AEZS 108 and AEZS 125. BMC Cancer. 14, 847-859 (2014). 7. Kakar, S. S., Jin, H., Hong, B., Eaton, J. W., Kang, K. A. LHRH receptor targeted therapy for breast cancer. Adv Exp Med Biol. 614, 285-296 (2008). 8. Limonta, P. et al. GnRH Receptors in Cancer: From Cell Biology to Novel Targeted Therapeutic Strategies. Endocr Rev. 33, 784–811 (2012). 9. Cook, T., Sheridan, W. P. Development of GnRH Antagonists for Prostate Cancer: New Approaches to Treatment. The Oncologist. 5, 162-168 (2000). 10. Shore, N. D., Abrahamsson, P. A., Anderson, J., Crawfors, E. D., Lange, P. New Considerations for ADT in Advanced Prostate Cancer and the Emerging Role of GnRH Antagonists. Prostate Cancer Prostatic Dis. 16, 7-15 (2013). 11. Liu, S. V. et al. Expression of Receptors for Luteinizing Hormone-Releasing Hormone (LH-RH) in Prostate Cancers following Therapy with LH-RH Agonists. Clin Cancer Res. 16, 4675-4680 (2010). 12. Nagy, A., Schally, A. V. Targeting of Cytotoxic Luteinizing Hormone-Releasing Hormone Analogs to Breast, Ovarian, Endometrial, and Prostate Cancers. Biol Reprod. 73, 851-859 (2005). 13. Reutter, M., Emons, G., Gründker, C. Starving tumors: inhibition of glycolysis reduces viability of human endometrial and ovarian cancer cells and enhances antitumor efficacy of GnRH receptor-targeted therapies. Int J Gynecol Cancer. 23, 34-40 (2013). 14. Engel, J. B. et al. Effective treatment of experimental human endometrial cancers with targeted cytotoxic luteinizing hormone-releasing hormone analogues AN-152 and AN207. Fertil Steril. 83, 1125-1133 (2005). 15. Cheung, L. W., Yung, S., Chan, T. M., Leung, P. C., Wong, A. S. Targeting Gonadotropin-Modulating Tumor-mesothelial Adhesion. Mol Ther. 21, 78-90 (2013). 16. Gründker, C., Nia, A H., Emons, G. Gonadotropin-releasing hormone receptor–targeted gene therapy of gynecologic cancers. Mol Cancer Ther. 4, 225-231 (2005).

15 ACS Paragon Plus Environment

Bioconjugate Chemistry 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

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

17. Engel, J. B. et al. Targeted chemotherapy of endometrial, ovarian and breast cancers with cytotoxic analogs of luteinizing hormone-releasing hormone (LHRH). Arch Gynecol Obstet. 286, 437-442 (2012). 18. Gründker, E. J., Ernst, J., Reutter, M. D., Ghadimi, B. M., Emons, G. Effective targeted chemotherapy using AEZS-108 (AN-152) for LHRH receptor-positive pancreatic cancers. Oncol Rep. 26, 629-635 (2011). 19. Szepeshazi, K. et al. Powerful Inhibition of Experimental Human Pancreatic Cancers by Receptor Targeted Cytotoxic LH-RH analog AEZS-108. Oncotarget. 4, 751-760 (2013). 20. Treszl A, et al. Substantial expression of luteinizing hormone-releasing hormone (LHRH) receptor type I in human uveal melanoma. Oncotarget. 4, 1721-1728 (2013). 21. Jaszberenyi, M. et al. Inhibition of U-87 MG glioblastoma by AN-152 (AEZS-108), a targeted cytotoxic analog of luteinizing hormone-releasing hormone. Oncotarget. 4, 422432 (2013). 22. Curtis, K. K. et al. Novel LHRH‑receptor‑targeted cytolytic peptide, EP‑100: first‑in‑human phase I study in patients with advanced LHRH‑receptor‑expressing solid tumors. Cancer Chemother Pharmacol. 73, 931–941, (2014). 23. Wen, D. et al. LHRH-Conjugated Micelles for Targeted Delivery of Antiandrogen to Treat Advanced Prostate Cancer. Pharm Res. 31, 2784–2795 (2014). 24. Nukolova, N. V. et al. LHRH-targeted nanogels as a delivery system for cisplatin to ovarian cancer. Mol Pharm. 10, 3913-3921 (2013). 25. Aicher, B. et al. LHRH receptor targeting as mechanism of anti-tumor activity for cytotoxic conjugates of Disorazol Z with the LHRH receptor agonistic peptide D-Lys6LHRH. AACR annual meeting, Washington D.C. Abstract #5476, (2013). 26. Karampelas, T. et al. GnRH-Gemcitabine conjugates for the treatment of androgenindependent prostate cancer: pharmacokinetic enhancements combined with targeted drug delivery. Bioconjugate Chem. 16, 813-823 (2014). 27. Liu, H. et al. Ultrasound-Mediated Destruction of LHRHa-Targeted and PaclitaxelLoaded Lipid Microbubbles Induces Proliferation Inhibition and Apoptosis in Ovarian Cancer Cells. Mol Pharm. 11, 49–58 (2014). 28. Vegt, E. et al. Renal uptake of different radiolabelled peptides is mediated by megalin: SPECT and biodistribution studies in megalin-deficient mice. Eur J Nucl Med Mol Imaging. 38, 623–632 (2011). 29. Hosseinimehr, S. J., Tolmachev, V., Orlova, A. Liver uptake of radiolabeled targeting proteins and peptides: considerations for targeting peptide conjugate design. Drug Discov Today. 21/22, 1224-12232 (2012). 30. Tucci, F. C. et al. 3-[(2R)-Amino-2-phenylethyl]-1-(2,6-difluorobenzyl)-5-(2-fluoro-3methoxyphenyl)-6-methylpyrimidin-2,4-dione (NBI 42902) as a Potent and Orally Active Antagonist of the Human Gonadotropin-Releasing Hormone Receptor. Design, Synthesis, and in Vitro and in Vivo Characterization. J Med Chem. 48, 1169-1178 (2005). 31. Hao, D., Sun, L., Hu, X., & Hao, X. 99mTc-LHRH in tumor receptor imaging. Oncol. lett, 14, 569-578 (2017).

16 ACS Paragon Plus Environment

Page 16 of 18

Page 17 of 18 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

451 452 453 454 455 456 457 458 459 460 461

Bioconjugate Chemistry

32. Li, M., Tang, Z., Zhang, Y., Lv, S., Li, Q. and Chen, X. Targeted delivery of cisplatin by LHRH-peptide conjugated dextran nanoparticles suppresses breast cancer growth and metastasis. Acta biomaterialia, 18, 132-143 (2015). 33. Raffaele, C. et al. Total Synthesis and Biological Evaluation of Tubulysin Analogues. J Org Chem. 81, 10302-10320 (2016). 34. Srinivasarao, M., Galliford, C.V. and Low, P.S. Principles in the design of ligandtargeted cancer therapeutics and imaging agents. Nat Rev Drug Discov. 14, 203-219 (2015). 35. Srinivasarao, M. and Low, P.S. Ligand-Targeted Drug Delivery. Chem Rev. 117, 1213312164 (2017).

462 463

17 ACS Paragon Plus Environment

Bioconjugate Chemistry 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

306x111mm (149 x 149 DPI)

ACS Paragon Plus Environment

Page 18 of 18