ChAcNLS, a Novel Modification to Antibody-Conjugates Permitting

Apr 25, 2016 - Michel PaquetteSimon BeaudoinMylene-Annie TremblaySteve JeanAngel ... Nadia Ekindi-Ndongo , Pierre-Luc Boudreaut , Marc-Andre Bonin ...
0 downloads 0 Views 3MB Size
Subscriber access provided by BALL STATE UNIV

Article

ChAcNLS, a novel modification to antibody-conjugates permitting target cell-specific endosomal escape, localization to the nucleus and enhanced total intracellular accumulation Simon Beaudoin, Andreanne Rondeau, Olivier Martel, MarcAndre Bonin, Johan E van Lier, and Jeffrey V. Leyton Mol. Pharmaceutics, Just Accepted Manuscript • DOI: 10.1021/acs.molpharmaceut.6b00075 • Publication Date (Web): 25 Apr 2016 Downloaded from http://pubs.acs.org on April 26, 2016

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 free 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 accessible to all readers and 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.

Molecular Pharmaceutics 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 34

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

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

Molecular Pharmaceutics

ChAcNLS, a novel modification to antibody-conjugates permitting target cell-specific endosomal escape, localization to the nucleus and enhanced total intracellular accumulation Simon Beaudoina, Andreanne Rondeaua, Olivier Martela, Marc-Andre Boninb, Johan E. van Liera,c, and Jeffrey V. Leytona,c* a

Departément de médecine nucléaire et radiobiologie, bPlateforme de synthèse de peptides et de sondes d'imageries, Faculté de médecine et sciences de la santé, Université de Sherbrooke and c Centre de’Imagerie Moléculaire de Sherbrooke (CIMS), 3001 12e Avenue Nord, Sherbrooke, Québec, Canada J1H5N4 *Address correspondence to: Jeffrey V. Leyton PhD Département de médecine nucléaire et radiobiologie Université de Sherbrooke 3001, 12e Avenue Nord, Sherbrooke, QC, Canada J1H5N4 Tel. (819) 820-6868; FAX: (819) 564-5442 E-mail: [email protected]

Keywords: nuclear localization, endosome escape, intracellular accumulation, antibody-conjugates

1 ACS Paragon Plus Environment

Molecular Pharmaceutics

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

1

Page 2 of 34

Abstract

2

The design of antibody-conjugates (ACs) for delivering molecules for targeted applications in

3

humans has sufficiently progressed to demonstrate clinical efficacy in certain malignancies and

4

reduced systemic toxicity that occurs with standard non-targeted therapies. One area that can further

5

clinical success for ACs will be to increase their intracellular accumulation. However, entrapment

6

and degradation in the endosomal-lysosomal pathway, which ACs are reliant for the depositing of

7

their molecular payload inside target cells, leads to reduced intracellular accumulation. Innovative

8

approaches that can manipulate this pathway may provide a strategy for increasing accumulation. We

9

hypothesized that escape from entrapment inside the endosomal-lysosomal pathway and redirected

10

trafficking to the nucleus could be an effective approach to increase intracellular AC accumulation in

11

target cells. Cholic acid (ChAc) was coupled to the peptide CGYGPKKKRKVGG containing the

12

nuclear localization sequence (NLS) from SV-40 large T-antigen, termed ChAcNLS. ChAcNLS was

13

conjugated to the mAb 7G3 (7G3-ChAcNLS) that has nanomolar affinity for the cell-surface

14

leukemic antigen interleukin-3 receptor-α (IL-3Rα). Our aim was to determine whether 7G3-

15

ChAcNLS increased intracellular accumulation while retaining nanomolar affinity and IL-3Rα-

16

positive cell selectivity. Competition ELISA and cell treatment assays were performed. Cell

17

fractionation, confocal microscopy, flow cytometry, and Western blot techniques were used to

18

determine the level of antibody accumulation inside cells and in corresponding nuclei. In addition,

19

the radioisotope copper-64 (64Cu) was also utilized as a surrogate molecular cargo to evaluate

20

nuclear and intracellular accumulation by radioactivity counting. 7G3-ChAcNLS effectively escaped

21

endosome entrapment and degradation resulting in a unique intracellular distribution pattern. MAb

22

modification with ChAcNLS maintained 7G3 nanomolar affinity and produced high selectivity for

23

IL-3Rα-positive cells. In contrast, 7G3 ACs with the ability to either escape endosome entrapment or

24

traffic to the nucleus was not superior to 7G3-ChAcNLS for increasing intracellular accumulation.

25

Transportation of

26

intracellular radioactivity accumulation. Thus, ChAcNLS is a novel mAb functionalizing technology

27

that demonstrates its ability to increase AC intracellular accumulation in target cells through

28

escaping endosome entrapment coupled to nuclear trafficking.

64

Cu when complexed to 7G3-ChAcNLS also resulted in increased nuclear and

29 30 31 2 ACS Paragon Plus Environment

Page 3 of 34

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

1

Molecular Pharmaceutics

1. Introduction

2

Antibody-conjugates (ACs) have impacted the healthcare industry for their ability to deliver

3

attached molecular payloads, such as chemotherapeutics and radioisotopes, selectively against cancer

4

cells but new directions to advance AC efficacy must be sought to improve responses in tumor

5

regression and duration. The enhancement of AC intracellular retention in target cells is one area that

6

could increase their effectiveness.1 Upon receptor-mediated internalization, ACs are typically

7

trapped inside endosomes and trafficked through the endosomal-lysosomal pathway.2 Lysosomes are

8

membrane-enclosed organelles that contain an array of digestive enzymes and receive proteins

9

transported by endosomes through vesicle membrane fusion and results in the release of active drug

10

catabolites. Despite this method of targeted intracellular drug accumulation, most patients eventually

11

progress.3-6 Important factors that cause cancer cell resistance have been shown to be the

12

overexpression of multidrug resistant (MDRs) transporters and the down regulation of target receptor

13

cell surface levels for chemotherapeutics7, 8 and intracellular dehalogenation for radioiodinated ACs.9

14

Receptor recycling pathways and their increased use by cancer cells has also been implicated to

15

reduce the intracellular accumulation of the internalized AC.10 Therefore, avoiding entrapment in

16

these intracellular pathways is an important area to improve the cellular accumulation of transported

17

payloads and for maximizing AC activity.

18

The functionalization of monoclonal antibodies (mAbs) with cell-penetrating peptides has

19

resulted in remarkable increases in intracellular accumulation when cells are treated with these types

20

of ACs.11-16 However, this advancement in AC cellular accumulation has been mostly for allowing

21

mAbs to access and target specific molecules inside cells that would otherwise be unavailable for

22

antibodies to target. Of the few reports that attempt to utilize ACs equipped with cell-penetrating

23

peptides as therapeutic agents against cell surface cancer-specific receptors,17-20 all suffered from

24

high accumulation in non-target cells or tissues and thus limited in their application for targeted

25

delivery.

26

shown impressive abilities to escape endosomes and enter the cytoplasm while maintaining target

27

cell selectivity.18, 21 However, it is yet to be determined whether increased escape by these ACs

28

corresponds to an increase in intracellular accumulation.

Recent advancements whereby ACs functionalized with pH-sensitive polymers have

29

Another recent advancement has been to empower ACs to achieve multi-selective targeting

30

by attaching peptides that harbor compartment-localizing amino acids.22, 23 In particular to this study,

31

the nuclear localization signal (NLS) sequence from SV-40 Large T-antigen has previously been

3 ACS Paragon Plus Environment

Molecular Pharmaceutics

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 4 of 34

1

incorporated into synthetic peptides and conjugated to proteins and demonstrated the ability to direct

2

the transport of proteins into the nucleus.24 Although, the optimized NLS sequence is 25 amino acids

3

long,25 we have previously utilized the mAb, 7G3, conjugated to a 13-mer peptide

4

(CGYGPKKKRKVGG) harboring a segment of the NLS (underlined) sufficient for nuclear

5

translocation.26-30 7G3 is specific for the leukemic blasts and leukemic stem cells marker IL-3Rα.31,

6

32

7

cell selectivity.33 7G3-NLS was used to deliver the radioisotope cargo indium-111 (111In) inside the

8

nucleus. Molecular damage by

9

they travel only nanometer-micrometer distances they are more effective if delivered inside the

An advantage of this short sequence is that it does not penetrate cells and allows mAbs to maintain 111

In is due to its emissions of energetic Auger electrons.34 Because

10

nucleus.34 Unfortunately, cytotoxicity was not overwhelming relative to standard

111

11

evidence suggested it was due to ineffective nuclear localization caused by entrapment in the

12

endosomal-lysosomal and/or recycling pathways.27, 28, 30 This prompted our current efforts to develop

13

an AC that gains the ability to effectively escape endosome entrapment. Furthermore, we wanted to

14

determine if the combination of both endosome escape and nuclear trafficking enhances intracellular

15

accumulation in order to establish a unique innovation in AC design and support future studies

16

whereby ACs are equipped with cytotoxic molecular payloads and tested for improved therapeutic

17

activity.

In-7G3 and the

18

In this report, we present the initial characterization of the bile acid, cholic acid (ChAc)-NLS

19

fusion compound (ChAcNLS) conjugated to 7G3. In general, bile acids are known for their role in

20

dietary lipid breakdown.35 However, bile acids have been shown to be essential for viral escape from

21

endosomes into the cytoplasm through a non-detergent mechanism.36, 37 Non-enveloped viruses that

22

cannot rely on membrane fusion, recruit ChAc to trigger ceramide formation in endosome

23

membranes. Increased ceramide in the endosome forms channels or makes membrane flip-flop

24

sufficient for protein traversal.38-41 42 Although this mechanism is not completely resolved, this led us

25

to hypothesize that ChAc functionalization of 7G3 could allow for the selective disruption of

26

endosomal membranes and not plasma membranes. Coupled to NLS, ChAcNLS conjugation to 7G3

27

could create an AC with the additional ability to efficiently localize to the nucleus of cells with intact

28

receptor selectivity. As a proof-of-principle model system, we used TF-1a leukemic cells that

29

express IL-3Rα. We examined the selectivity of 7G3-ChAcNLS by comparing its treatment on IL-

30

3Rα-negative cells or by functionalizing isotype-matched non-specific mAbs with ChAcNLS. We

31

examined the efficiency of 7G3-ChAcNLS by comparing it to unmodified 7G3 and different 7G3 4 ACS Paragon Plus Environment

Page 5 of 34

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

Molecular Pharmaceutics

1

ACs with either limited endosome escape or nuclear translocation capabilities. Importantly, we

2

evaluated what were the effects of endosome escape coupled to nuclear translocation on intracellular

3

accumulation. For extending this technology to molecular payloads, we tested the TF-1a nuclear and

4

intracellular accumulation of transported radioisotope copper-64 (64Cu).

5 6

2. Materials and methods

7

2.1 Human Cells and mAbs

8 9

TF-1a leukemia and Raji Burkitt’s lymphoma cells were obtained from the American Type

10

Tissue Collection (Manassas, VA). TF-1a cells are positive for IL-3Rα by flow cytometry

11

(corresponding to 7.8 x 103 receptors/cell for 111In-labeled chimeric version of 7G3,43 and Raji cells

12

are IL-3Rα-negative. Cells were cultured in RPMI 1640 medium supplemented with 1%

13

penicillin/streptomycin, 1% non-essential amino acids, 1% sodium pyruvate, 10% heat inactivated

14

FBS, and 0.2% amphotericin B (Wisent, Quebec, Canada). 7G3 and isotype control mIgG2a were

15

purchased from BD Biosciences (Ontario, Canada).

16 17

2.2 7G3-ChAcNLS construction

18 19

ChAcNLS was designed with the nuclear localization sequence from SV-40 large T antigen

20

with GYG and GG residues at the N- and C-terminus as spacers, respectively (Fig. 1A). The N-

21

terminus was capped with a cysteine for conjugation to ChAc and 7G3. Controls included 7G3

22

conjugated with ChAc or NLS only and with compounds ChAc-CGYGPLKLRKVGG (ChAcLeu)

23

and ChAc-CGYGPDapKDapRKVGG (Dap=2,3-diaminopropionic acid; ChAcDap). Control

24

ChAcLeu was to conjugate 7G3 with a compound that enabled endosome escape but did not possess

25

a functional NLS but was of similar size. Control ChAcDap was used to conjugate 7G3 with a

26

compound that enabled endosome escape without NLS capabilities but possessed the same net

27

charge (+5) as NLS. 7G3 or IgG2a was first maleimide activated for conjugation to the N-terminal

28

cysteine of the peptide. Maleimide groups were introduced into 7G3 or IgG2a by reaction of 10

29

mg/mL 7G3 in PBS, pH 7.6, with 10-, 25-, or 50-fold molar excess of sulfosuccinimidyl-4-(N-

30

maleimidomethyl)-cyclohexane-1-carboxylate (sulfo-SMCC; VWR, Quebec, Canada) at room

31

temperature for 1 h. Maleimide-derivatized mAbs were purified on a Sephadex G-50 (Sigma5 ACS Paragon Plus Environment

Molecular Pharmaceutics

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 6 of 34

1

Aldrich) column eluted with PBS, pH 7.0. Fractions containing maleimide-7G3 or –IgG2a were

2

transferred to a Centricon YM-100 ultrafiltration device (EMD Millipore, Ontario, Canada) and

3

concentrated to 10 mg/mL, which was then reacted with 100-fold molar excess of ChAcNLS or

4

control compounds for 18 h at 4°C. ACs were purified from excess peptide and concentrated in PBS,

5

pH 7.4, by ultrafiltration through a Centricon-YM-100. For peptide synthesis and 7G3-ChAcNLS

6

characterizations please refer to Supporting Information.

7 8

2.3 Exposure of cells to mAb conjugates (Supplemental Fig. 1 in the Supporting Information)

9 10

Unless indicated, 5 x 106 TF-1a cells were exposed to 200 nmol/L of ACs in RPMI/10%FBS

11

for 1 h at 37°C. Cells were then washed 3x in ice-cold PBS and suspended in RPMI/10%FBS for an

12

additional 1 h at 37°C as a post-incubation. This allowed for the evaluation of internalization and

13

intracellular accumulation of the ACs to be monitored by the methods utilized in this study. Cells

14

were then centrifuged and washed in ice-cold PBS for further processing. Assays were performed in

15

triplicate and repeated a minimum of three times.

16 17

2.4 7G3-ChAcNLS target cell selectivity by confocal microscopy

18 19

To determine 7G3-ChAcNLS selectivity for IL-3Rα-positive TF-1a cells, cells were

20

suspended in 0.1 mL of PBS containing 0.25% Trypsin (Wisent Bio Products, Quebec, Canada) and

21

0.25% EDTA and incubated at 37°C for 3 min, which was used to remove surface bound ACs.

22

Trypsin was neutralized with the addition of 0.4 mL of RPMI/10% FBS. Very similar trypsin

23

procedures have previously been reported.44-46 Trypsinized cells were fixed in 1% paraformaldehyde/

24

1% sucrose on ice for 30 min. Cells were then washed and permeabilized with 0.15% Triton X for 5

25

min. Cells were then washed and suspended in 0.5 mL PBS containing 2 µg/mL of anti-murine Fc

26

secondary polyclonal antibody conjugated to Alexa Fluor 647 (AF647; Life technologies, Ontario,

27

Canada) for 1 h at room temperature in the dark. In addition, to control for potential non-specific

28

trafficking activity by the positive charged ChAcNLS on fixed cells, non-treated TF-1a cells were

29

also fixed and permeabilized. The fixed and permeabilized TF-1a cells were post-treated with

30

ChAcNLS-7G3 for 1 h at 37°C followed by washing and staining with AF647 antibodies. Cells were

31

stained with propidium iodide (PI) 5 min prior to analysis. For confocal microscopy, cells were 6 ACS Paragon Plus Environment

Page 7 of 34

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

Molecular Pharmaceutics

1

mounted onto glass slides with SlowFade mounting media (Life technologies) and covered with a

2

glass coverslip. All images were acquired on a FV1000 scanning confocal microscope (Olympus,

3

Tokyo, Japan) coupled to an inverted microscope using a 63x oil immersion objective. PI

4

fluorescence was detected with a 488 nm argon laser and a spectral scanning prism fixed for 600-650

5

nm. AF647 fluorescence was detected using a 633 nm helium-neon laser and a spectral scanning

6

prism fixed for 650-700 nm. Fluorescence emissions from PI and AF647 were collected sequentially.

7

Serial horizontal optical sections of 1024 x 1024 pixels with 2-times line averaging were taken at 0.5

8

µm intervals through the entire cell thickness. Images were acquired in the same day with identical

9

instrument settings. Images are presented as the stacked z-projections (maximum intensity) from 4

10

consecutive slices. Each slice was focused for maximum intensity without photobleaching. The

11

consequence is that low fluorescence intensity is not captured causing some cells to appear negative.

12

Images were pseudocolored and merged (FluoView; Olympus). The ability of the 7G3 conjugates to

13

accumulate inside cells was determined by counting the proportion of AF647 fluorescence positive

14

cells and visually analyzing fluorescence intensity.

15 16

2.5 Intracellular accumulation by flow cytometry and Western blot

17 18

Treated cells were fixed and permeabilized and incubated with AF647 as described in section

19

2.4. AC accumulation was calculated as the geometric mean fluorescence intensity (MFI) normalized

20

to background MFI levels within the intact fixed and permeabilized cell population (as determined

21

by the forward scatter/side scatter distribution47 and positive PI staining) incubated with AF647-only

22

as determined by CellQuest Pro software (BD Biosciences).

23

For Western Blot analysis, after trypsin neutralization, cells were washed with PBS and lysed

24

in RIPA buffer. Total protein concentration was determined using the BCA Protein Assay Reagent

25

(VWR). 10 µg of protein were loaded into wells in a 12% SDS gel and electrophoresed at 180 V for

26

1 h. The gel was transferred on to a PVDF membrane at 100 V for 1 h. PVDF membranes were then

27

washed in TBS/0.1% Tween 20 and placed into a 5% milk/TBS/0.1%Tween 20 blocking solution for

28

1 h followed by washing 3x in TBS/0.1% Tween 20. Membranes were then incubated in 2.5%

29

milk/PBS containing 1 µg/mL of polyclonal rabbit antibodies against murine Fc conjugated to

30

horseradish peroxidase (HRP; Life technologies). HRP signal was then developed by enhanced

7 ACS Paragon Plus Environment

Molecular Pharmaceutics

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 8 of 34

1

chemiluminescence (BioRad). Probing for anti-Actin (Sigma-Aldrich) as the loading control was

2

performed under similar procedures.

3 4

2.6 Nuclear fractionation and enrichment for analyzing nuclear localization and accumulation

5

(Suppl. Fig. 1)

6 7

To isolate the cell nuclei, cells were washed in ice-cold PBS and then lysed by suspending in

8

0.05% NP-40, 10 mmol/L Tris pH 7.5, 10 mmol/L NaCl, 3 mmol/L MgCl2 buffer on ice for 10 min.

9

Lysed cells were then centrifuged at 80g for 5 min. The nuclei pellet was then washed without NP-40

10

and centrifuged at 80g for an additional 5 min. Nuclei were washed 3x in ice cold PBS. The degree

11

of isolated nuclei enrichment was determined by Western Blot and flow cytometry. Western Blots

12

were performed as previously described with the exception that mAbs used were specific for the

13

well-established nuclear marker lamin A/C (Santa Cruz Biotechnology Inc., Dallas, TX) or plasma

14

membrane/late-endosome/lysosomal marker Lamp 1 (Sigma-Aldrich). These two markers ensure

15

nuclei are free of contaminants from the plasma membrane, late-endosomes or lysosomes and for

16

loading control and analysis by densitometry. Membranes were washed and developed as previously

17

described. For flow cytometric analysis, changes in FSC vs SSC from intact cells and in isolated

18

nuclei were compared (Suppl. Fig. 2). The smaller nuclei population is distinct from intact cells.

19

Nuclei were stained with PI, which binds to DNA to confirm DNA is contained in the nuclei

20

population. Nuclei were then processed and analyzed by confocal microscopy and flow cytometry to

21

determine nuclear localization and accumulation, respectively, as previously described for intact

22

cells.

23 24

2.6 Radioactivity cargo studies

25 26

Approximately 1.5 mg of 7G3 (5 µg/µL in 50 mM NaHCO3 buffer, pH 7.5) was reacted with

27

1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA; for complexing copper-64 (64Cu)).

28

DOTA-conjugates were purified on an Amicon Ultra centrifugal filter (Millipore). Approximately

29

500 µg of DOTA-conjugates were reacted with sulfo-SMCC and conjugated to ChAcNLS or NLS as

30

previously described and then purified by centrifugation. 50 µg of DOTA-ChAcNLS-conjugates

31

were placed in 50 µL of 0.1 M ammonium acetate, pH 5.5 for 1 h at room temperature with 8 MBq

8 ACS Paragon Plus Environment

Page 9 of 34

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

Molecular Pharmaceutics

1

of 64CuCl2. The radioimmunoconjugates were purified by dilution in PBC followed by centrifugation

2

in Amicon centrifugal filters. Radiochemical purity was determined by instant thin layer

3

chromatography developed in 100 mM sodium citrate, pH 5.5. The final radiochemical purity of all

4

radioimmunoconjugates was >98%. 100 nmol/L of radioimmunoconjugates were added to 4 x 106

5

TF-1a cells and incubated as described in section 2.3. Cells were fractionated as described in section

6

2.4 and nuclear radioactivity in each fraction measured in a gamma counter and reported as counts

7

per min (CPM) per fraction. We considered intracellular accumulation as the product of the

8

radioactivity in nuclear and cytoplasmic fractions.

9 10

2.7 Statistical analysis

11 12

Data are presented as the mean ± SD. Significant differences were tested using an unpaired,

13

two-sided Student’s t-test (p