Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Sep 18.
Published in final edited form as: Bioconjug Chem. 2022 Apr 7;33(4):718–725. doi: 10.1021/acs.bioconjchem.2c00083

Cyanine Masking: A Strategy to Test Functional Group Effects on Antibody Conjugate Targeting

Ek Raj Thapaliya 1, Syed Muhammad Usama 2, Nimit L Patel 3, Yang Feng 4, Joseph D Kalen 5, Brad St Croix 6, Martin J Schnermann 7
PMCID: PMC10506421  NIHMSID: NIHMS1925989  PMID: 35389618

Abstract

Conjugates of small molecules and antibodies are broadly employed diagnostic and therapeutic agents. Appending a small molecule to an antibody often significantly impacts the properties of the resulting conjugate. Here, we detail a systematic study investigating the effect of various functional groups on the properties of antibody–fluorophore conjugates. This was done through the preparation and analysis of a series of masked heptamethine cyanines (CyMasks)-bearing amides with varied functional groups. These were designed to exhibit a broad range of physical properties, and include hydrophobic (−NMe2), pegylated (NH-PEG-8 or NH-PEG-24), cationic (NH-(CH2)2NMe3+), anionic (NH-(CH2)2SO3), and zwitterionic (N-(CH2)2NMe3+)-(CH2)3SO3) variants. The CyMask series was appended to monoclonal antibodies (mAbs) and analyzed for the effects on tumor targeting, clearance, and non-specific organ uptake. Among the series, zwitterionic and pegylated dye conjugates had the highest tumor-to-background ratio (TBR) and a low liver-to-background ratio. By contrast, the cationic and zwitterionic probes had high tumor signal and high TBR, although the latter also exhibited an elevated liver-to-background ratio (LBR). Overall, these studies provide a strategy to test the functional group effects and suggest that zwitterionic substituents possess an optimal combination of high tumor signal, TBR, and low LBR. These results suggest an appealing strategy to mask hydrophobic payloads, with the potential to improve the properties of bioconjugates in vivo.

Graphical Abstract

graphic file with name nihms-1925989-f0001.jpg

INTRODUCTION

Conjugates of monoclonal antibodies (mAbs) and small molecules are indispensable tools for diagnostic and therapeutic applications. While long used for various cellular imaging applications (i.e., immunolabeling), the past decade has seen significant progress in the translation of mAb conjugates into clinical settings. For example, conjugates of fluorescent molecules and mAbs have emerged as powerful tumor imaging agents for fluorescence-guided surgery.1,2 In the context of therapeutics, antibody drug conjugates (ADCs) harness the specificity of antibodies to selectively deliver potent payload molecules.3 Despite significant progress validating these approaches, significant challenges remain. In particular, many small molecule payloads interfere with the intrinsic targeting and clearance of the parent antibody.46 Various strategies are being explored to circumvent this issue. These include homogenous labeling strategies, which have been explored extensively for ADCs. While such strategies improve certain in vivo properties, they increase the complexity of the labeling process.79 For optical probes, the use of highly charged, persulfonated fluorophores is a long established strategy designed to address the issue of fluorophore aggregation on the protein surface.10 This strategy led to the creation of numerous broadly used fluorophores, including the “Cy” and “Alexa Fluor” series. While these probes are optimized for in vitro efforts, more recent results from our group and others have shown that the highly charged, but net neutral (i.e. zwitterionic) fluorophores can improve the targeting of mAbs in vitro and in vivo.4,16

A critical, still outstanding question is what functional group can best mitigate the impact of small molecule labeling on mAb targeting. Here, we pursue a strategy centered on comparing antibody conjugates of a series of modified near-infrared (NIR) fluorescent dyes. While intrinsically hydrophobic, these molecules can be synthetically modified with polar functional groups. Several prior studies, including those from our group, have examined the impact of modifications on the in vivo properties of mAb–fluorophore conjugates.1121 While these prior studies provided key insights, they are not systematic from a “functional group” perspective making it hard to draw larger lessons (see Figure S1 for previous molecules). Here, we address this issue with single-point alterations to substituents distal to the cyanine scaffold. We hypothesize that this approach will provide insights that can broadly inform the design of various bioconjugates.

Below, we detail the synthesis and evaluation of a series of masked heptamethine cyanines (CyMasks) and their antibody conjugates. These probes enable the unbiased assessment of chemical masking strategies on tumor targeting and off-target accumulation of mAb conjugates. In line with prior results, we find that the most hydrophobic probe exhibits the most non-selective in vitro uptake and the lowest in vivo tumor targeting. We also find that anionic and cationic masking groups lead to increased dye aggregation and reduced tumor signals. Notably, pegylated probes, which have been broadly applied in other contexts,2227 exhibit excellent in vitro uptake but reduced in vivo tumor targeting. By contrast, zwitterionic probes, bearing both quaternary ammonium and sulfonate functional groups, exhibit excellent in vitro and in vivo properties. These results suggest that the combination of highly charged, but net neutral, substituents may offer significantly improved biophysical properties with implications to a range of bioconjugates.

RESULTS AND DISCUSSION

Synthesis and Characterization of CyMask and mAb Conjugates.

In designing the CyMask series, we sought to examine a range of polar substituents and chose representative zwitterionic, cationic, anionic, pegylated, and hydrophobic functional groups. Specifically, we synthesized six CyMask dyes containing amides substituted with PEG-8, PEG-24, N-ethyl-sulfonate (Sulfo), N-ethyl-trimethyl-ammonium (Quat), N-trimethyl-propyl-ammonium-N-butyl-sulfonate (Zwit), and a hydrophobic derivative, methylamine (Me). As shown in Scheme 1, the CyMask-NHS esters were synthesized from the known precursor (1) using our previously reported Smiles rearrangement method.11 Compound 1 underwent substitution with N-methyl-ethanolamine in MeCN at 80 °C to obtain 2. The subsequent Smiles rearrangement (C4′-N to C4′-O) reaction was carried out using an initial TFA-induced rearrangement followed by electrophilic trapping in THF in the presence of NaHCO3 at 50 °C to give methyl ester 3. This common precursor 3 was used to couple with primary amines 4a–f to give amides 5a–f, and subsequent saponification with LiOH provided the free carboxylic acids (6a–f). We compared the photophysical properties of all six CyMask carboxylic acids (6a–f) and found similar optical properties in PBS pH 7.4 and 10% FBS (Table 1 and Figures S2 and S3) across this series and relative to other C4′-O-linked heptamethine cyanines.1116 These observations confirm the minimal impact of the distal modification on the optical properties of the free molecules.

Scheme 1.

Scheme 1.

Synthesis of Compounds in CyMask Series and Conjugation with Panitumumab

Table 1.

Key Spectroscopic Properties

CyMask-CO2H λmax,abs (nm) λmax,em (nm) ΦF εa (M−1 cm−1) brightness (ε × ΦF) net charge molecular weight
CyMask-Me-CO2H (6a) 766 781 0.14 161,000 22,500 0 779.0
CyMask-PEG-8-CO2H (6b) 766 781 0.13 203,000 26,400 0 1483.9
CyMask-PEG-24-COOH (6c) 766 781 0.15 182,300 27,500 0 2893.6
CyMask-Sulfo-CO2H (6d) 764 780 0.11 174,000 19,000 -2 965.4
CyMask-Quat-CO2H (6e) 764 780 0.12 184,000 22,000 +2 923.3
CyMask-Zwit-CO2H (6f) 764 780 0.11 184,000 20,240 0 1165.5
a

Molar absorption coefficients of all dyes were measured in 1:1 MeOH-PBS pH 7.4.

We then examined the impact of the masking functional group on the properties of mAb conjugates. The carboxylic acids 6a–f (CyMask-CO2H) were converted to CyMask-NHS esters (7a–f) using N,N,N′,N′-tetramethyl-O-(N-succinimidyl) uronium tetrafluoroborate (TSTU) and were used in the bioconjugation step. We choose to first generate conjugates with panitumumab (Pan), an anti-EGFR FDA-approved monoclonal antibody, which has been used extensively for imaging applications.2,1116 All six NHS esters were reacted with panitumumab (Pan) in 50 mM PBS at pH 7.4 with molar excesses of 3, 6, and 10 equiv to obtain the lysine-labeled Pan conjugates with degrees of labeling (DOL) of 1, 2, and 3 (±0.3), respectively. Two purification methods were applied to the remove unlabeled probe. The dye conjugates were passed through a spin desalting column and stored overnight at 4 °C to dissociate non-covalently bound free dyes. The resulting solutions were purified a second time by passing through a desalting column eluting with pH 7.4 PBS. All conjugates provided homogenous solutions with the stable absorption spectra over a several-month timeframe.

The photophysical properties of these mAb-CyMask conjugates were analyzed in detail. The absorbance spectra were recorded in PBS pH 7.4 (Figure 1). While displaying nearly identical absorbance maxima, significant variation in the H-aggregate peak at 700 nm (Figure 1a insert) was observed across the various conjugates in PBS, but not in denaturing conditions (1:1 PBS:MeOH, Figure S5). This peak, which results from cyanine dimerization on the protein surface, is a well characterized consequence of protein labeling.2830 The magnitude of the H-aggregate was found to be dependent on both the probe and the DOL.3133 As a convenient means to quantify these effects, we examined the ratio of the absorbance values at 766 (monomer) and 700 (dimer) nm (Figure 1ad). Across the series, the values decrease along series Sulfo > Me > Zwit > PEG-8 > Quat > PEG-24. Notably, PEG-24 provided almost complete inhibition of H-aggregate formation, as noted elsewhere.23,34 The observation that sulfonated substituents led to the most substantial H-aggregate formation is somewhat surprising. Prior results clearly indicate that sulfonates appended to the indolenine heterocycles improve the properties of the resulting mAb conjugates.29,30,35,36 However, these results suggest that distal sulfonates are not sufficient to inhibit H-aggregation, and may even induce formation. Lastly, we evaluated the quantum yield (ΦF) of the mAb conjugates at DOL 3. Notably, the ΦF of thee conjugates did not vary dramatically (from 4.8 to 7.5%) in pH 7.4 PBS (Figure 1e). Additionally, we observe little correlation between the ΦF and either H-aggregation or the in vivo results described below. These observations suggest that the photon output of the fluorophore–mAb conjugates is relatively insensitive to the CyMask functional group, which simplifies the analysis of the in vivo data reported below.

Figure 1.

Figure 1.

Characterization of Pan-CyMask conjugates. (a) Normalized absorption of Pan-CyMask conjugates (DOL 3, pH 7.4 PBS). A zoomed-in view is shown in the inset to highlight the monomer and H-aggregated peaks. (b) Normalized absorption of Pan-CyMask conjugates (DOL 2, pH 7.4 PBS). (c) Normalized absorption spectra of Pan-CyMask (DOL 1, pH 7.4 PBS). (d) Aggregation ratio (absorbance ratio at 700/766 nm) of Pan-CyMask conjugates at DOL 1, 2, and 3 (pH 7.4 PBS) calculated from the absorption spectra. (e) ΦF (measured by a relative method) of Pan-CyMask conjugates (DOL 3, pH 7.2 PBS).

Comparison of Panitumumab-CyMask Conjugates.

We sought to characterize the impact of the CyMask modifications on tumor targeting and biodistribution of these agents. These studies were carried out using athymic nude female mice bearing EGFR+ MDA-MB-468 xenografts implanted subcutaneously in the right flank (5 × 106 cells, n = 5 per group).16 The mice were injected with 50 μg of the Pan-CyMask conjugates (DOL 3). Fluorescence images were obtained using an IVIS imaging system before injection and at 4, 24, 48, 72, and 168 h p.i. As early as 4 h, all the conjugates showed a significant tumor signal, which increased over the next 7 days (Figure 2a and Figures S6 and S7). After 7 days, the highest radiance fluorescence intensity was observed with the Zwit and Quat conjugates, with values that are ~5.5 and 5.0-fold higher compared to the Me conjugate, respectively. Analysis of the tumor-to-background ratio (TBR) indicates a rank order of Zwit ≈ Quat > PEG-24 > Sulfo ≈ PEG-8 ≈ Me 1 week p.i. (Figure 2b,c). While the fluorescence signal in most healthy organs remain low, significant background uptake was observed in liver, specifically for cationic and anionic dye conjugates, with the strongest signal at the 4 h time point (Figure 2d and Figures S6 and S7). We note that for PanCyMask-PEG-8 and -PEG-24, we observe an apparent bladder signal at 4 h p.i. (Figure S6). This signal may reflect a small amount of non-covalently bound dye that remained following purification or potentially some other degradation pathway. Of note, the in vivo imaging data were confirmed in a subsequent ex vivo study using Pan-CyMask-Zwit and Pan-CyMask-Me conjugates, where mice (n = 3 per group) were analyzed ex vivo at 48 h p.i. (Figure S8). Ex vivo histopathological analysis of tumor slices injected with Pan-CyMask-Zwit and Pan-CyMask-Me showed a heterogenous near-infrared signal throughout the tumor with the highest signal around vasculature (Figure 2e and Figure S9). In the tissue samples, we observed approximately twice the number of clearly labeled cells with Pan-CyMask-Zwit (~20–30%) as compared to Pan-CyMask-Me (~10–20%). Overall, these results indicate that the zwitterionic conjugates outperform the charged conjugates (Sulfo and Quat) with respect to liver uptake. Additionally, the zwitterionic conjugates improved mAb tumor uptake relative to hydrophobic, anionic, and pegylated conjugates.

Figure 2.

Figure 2.

In vivo and ex vivo fluorescence imaging of MDA-MB-468 xenograft tumor-bearing mice intravenously injected with Pan-CyMask conjugates (50 μg, DOL 3). (a) In vivo fluorescence images (right dorsum) at pre-injection and 4 h, 2 days, and 7 days post injection (p.i.). (b) Tumor signal (total radiant efficiency normalized to tumor size) after 7 days, (c) the tumor-to-background ratio after 7 days, and (d) the liver-to-background ratio after 4 h. Statistical analysis was performed using the Student’s t test. * p-value ≤ 0.05, *** p-value ≤ 0.001. (e) Representative histopathological images of tumor tissue sections (48 h, post injection) with Pan-CyMask-Me and Pan-CyMask-Zwit. The image is pseudo-colored: pink (near-infrared channel; Cy7), blue (DAPI), and green (autofluorescence).

We then studied the cellular uptake of these fluorophore conjugates in EGFR+ (MDA-MB-468) or EGFR– (MCF-7) cell lines. Epifluorescence microscopy demonstrated the high specific uptake of the CyMask conjugatesin MDA-MB-468 cells compared to the control cell line MCF-7 when incubated with the same concentration of Pan-CyMask conjugates and incubation time (250 μg/mL, 7 h) (Figure 3a). This approach indicates similar cellular uptake of all six Pan-CyMask conjugates. We quantified these differences in more detail by determining an apparent binding constant (EC50) using flow cytometry for three of the conjugates. The Pan-CyMask-PEG-24 conjugate showed slightly lower binding affinity (EC50 = 7.6 nM) compared to the Pan-CyMask-Zwit (EC50 = 4.5 nM) and Pan-CyMask-Me (EC50 = 6.9 nM) (Figure 3b). Overall, these data studies suggest modest differences in in vitro cellular uptake between the CyMask series and suggest that in vivo differences are largely due to altered clearance.

Figure 3.

Figure 3.

In vitro studies of Pan-CyMask conjugates in MDA-MB-468 (EGFR+), MCF-7 (EGFR−), and Kupffer cells. (a) Epifluorescence imaging of MDA-MB-468 (top row) cells and MCF-7 (bottom row) after incubation with Pan-CyMask conjugates (250 μg/mL, 7 h, 37 °C). (b) Binding affinity of Pan-CyMask conjugates in MDA-MB-468 cells. Cells were incubated with Pan-CyMask conjugates (0.1–0.3 μM) at an ice bath (4 °C) for 1 h in pH 7.4 PBS (with 1 mg/mL BSA containing 0.01% sodium azide) and fixed using paraformaldehyde. Mean fluorescence intensity (MFI ± SD, n = 3) was measured by flow cytometry. An amount of ~10,000 cells were counted. (c) Non-specific uptake of mAb conjugates in Kupffer cells. Cells were incubated with Pan-CyMask conjugates (0.1 mg/mL) at 37 ° C for 24 h in serum-containing media; MFI ± SD (n = 3) was measured by flow cytometry. An amount of ~10,000 cells were counted.

Next, we investigated a potential hepatic clearance pathway using an in vitro cellular model. Prior work examining the uptake of ADCs and other conjugates has revealed a critical role for stellate macrophages, or Kupffer cells, in mediating the uptake and clearance of bioconjugates labeled with hydrophobic payloads.37,38 To study the effect of the masking group on dye conjugates with respect to the non-specific uptake, we examined the uptake of these dye conjugates in rat Kupffer cells using flow cytometry.38 After incubation of Pan-CyMask conjugates with rat Kupffer cells for 24 h, the most hydrophobic dye conjugate, Pan-CyMask-Me, exhibited increased uptake compared to other conjugates (Figure 3c). Notably, irrespective of the nature of the masking group, more hydrophilic dye conjugates showed similar non-specific uptake by Kupffer cells. These results are in line with the prior observations that suggest a role for Kupffer cells in the uptake of hydrophobic small-molecule–mAb conjugates but suggest that additional mechanisms may be at play for the high uptake of the positively charged Pan-CyMask-Quat conjugate in vivo.

Comparison of Anti-CD-276 mAb-CyMask Conjugates.

To test the generality of these observations, we applied CyMask with a fully human anti-CD276 (B7 homolog H3, B7H3) antibody that binds both mouse CD276 and human CD276 with similar affinity.39,40 This target is overexpressed in the cancer cells and tumor neovasculature of multiple solid tumor types, and this antibody has been applied to create potent ADCs.3944 For the in vivo studies, we chose to compare the effects of positive charge (Quat), no charge (Me), and net neutral charge (Zwit) CyMask on tumor targeting and clearance. The conjugates were injected intravenously into mice bearing 200–250 mm3 JIMT-1 breast tumors grown orthotopically in the mammary fat pad. The tumor fluorescence intensity and TBR was highest for m276-SL-CyMask-Zwit (10.2) after 7 days (Figure 4a,b and Figures S10 and S11). These studies provide additional evidence that zwitterionic masking can improve the properties of mAb conjugates.

Figure 4.

Figure 4.

(a) Quantification of tumor uptake of m276-SL-CyMask-Me, Quat, and Zwit conjugates (50 μg; DOL 3) in JIMT-1 tumors after 168 h. (b) Tumor-to-Background ratio at 168 h post injection. (c) IVIS images at 4 h, 2 days, and 7 days post injection. Tumors are highlighted in red dotted circles. Statistical analysis was performed between groups at 168 h. Data points are displayed as mean ± SD, and the p-values were evaluated by the Student’s t test. * p-value ≤ 0.05, ** p-value ≤ 0.01, *** p-value ≤ 0.001.

CONCLUSIONS

In the studies outlined above, we detail an unbiased comparison of charge masking functional groups on the in vivo targeting properties of mAb–fluorophore conjugates. These studies reveal that zwitterionic and cationic shielding outperforms pegylated, anionic, and hydrophobic conjugates with respect to tumor uptake. Furthermore, cationic shielding leads to the highest off-target liver uptake at early imaging time points. Notably, significant in vitro non-specific uptake by Kupffer cells occurs with the hydrophobic derivative but not the charged or pegylated conjugates. In total, these results suggest that the zwitterionic shielding groups may offer the optimal combination of excellent targeting and minimal off-target uptake. These results align with prior studies from our group and others, which found that zwitterionic heptamethine cyanines exhibit improved tumor uptake when compared to conventional persulfonated heptamethine cyanines.16,17,45

These observations complement prior work to optimize the in vivo properties of therapeutic protein and nano-material modalities. In particular, extensive efforts have investigated the impact of mAb sequence modifications on clearance and tumor targeting. The general theme to emerge from these efforts is that cationization and anionization, the introduction of positively and negatively charged amino acids, respectively, both increase the plasma clearance rate, albeit through different mechanisms.46 Cationization generally increases tissue uptake, which can be advantageous for solid tumor uptake, but also leads to increased uptake in off-target organs.47 By contrast, anionization decreases tissue uptake due to increased whole-body (including blood) clearance through enhanced hepatic clearance.46 In general, these studies have settled on the notion that the isoelectric point (i.e., the pH at which the protein is net neutral) of native antibodies, which is typically between 7 and 8, is preferred. While efforts to chemically introduce charged functional groups, as we have done here, are rare, extensive efforts have examined the impact of pegylation strategies.23,34,4751 While offering significant benefits with respect to plasma half-life, this approach increases the size of protein significantly with consequent impacts on binding interactions.5255 Additionally, particularly when appended to long half-life proteins, such as mAbs, there is a potential for antibody-mediated immune responses.5658

Complementing efforts to augment the properties of proteins, the nanomaterials community has extensively studied the impact of charge and polarity as passivation strategies to improve the properties of otherwise hydrophobic species. In this context, there is significant evidence that zwitterionic functional groups reduce non-specific adsorption at the particle/liquid interface.37,59 The resulting decrease in non-specific interactions leads to reduced off-target uptake in various contexts. While, the exact mechanism of this effect is not entirely clear, previous studies have suggested a combination of charge shielding, minimization of membrane penetration, and protein stabilization.60

Here we introduce a series of optical probes that enable the quantitative assessment of functional group effects on mAb targeting in vivo. These results suggest that zwitterionic functional groups, which are highly charged, but net neutral modifications, may represent an important addition to the mAb-bioconjugate landscape. This strategy, which maintains the original charge profile of the parent protein, while simultaneously introducing a highly charged functional group, may represent a promising approach to mask the undesirable hydrophobicity of drug conjugates in the circulation. Future studies will investigate the potential impact of these modifications in homogeneous mAb conjugates, where we hypothesize that charge will also play a significant role.61,62 Additionally, we suggest that the incorporation of zwitterionic functional groups into otherwise hydrophobic therapeutic payloads may have significant promise. Studies toward these goals are underway in our group and will be reported in due course.

Supplementary Material

35389618_Schnermann_SM

ACKNOWLEDGMENTS

This work was supported by the Intramural Research Program of the National Institutes of Health (NIH), NCI-CCR. It was also supported by an NCI CCR FLEX Program Synergy Award (to M.S. and B.S.C.) and a Congressionally Directed Medical Research Program (CDRMP) Breast Cancer Research Program grant (award number W81WXH21-1-0109). We acknowledge Dr. James A. Kelley (National Cancer Institute) for providing the high-resolution mass spectrometry analysis. We thank Dr. Gary T. Pauly (National Cancer Institute) for assisting with LC/MS and HPLC purification. The Biophysics Resource, CCR is acknowledged for use of instrumentation. We would also like to thank Dr. Jeff Carrell (CCR-Frederick Flow Cytometry Core Laboratory) for assisting with flow cytometry and Dr. Elijah F. Edmondson (Molecule Histopathology Laboratory, Leidos Biomedical Research) for ex vivo analyzing histopathology samples.

Footnotes

The authors declare no competing financial interest.

ASSOCIATED CONTENT

Supporting Information

The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.bioconjchem.2c00083.

Synthesis procedures, characterization of CyMask probes, key intermediate compounds, photophysical properties of CyMask probes, details of in vitro and in vivo experiments, and supporting figures and tables (PDF)

Complete contact information is available at: https://pubs.acs.org/10.1021/acs.bioconjchem.2c00083

Contributor Information

Ek Raj Thapaliya, Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States.

Syed Muhammad Usama, Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States.

Nimit L. Patel, Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland 21702, United States

Yang Feng, Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute, NIH, Frederick, Maryland 21702, United States.

Joseph D. Kalen, Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland 21702, United States

Brad St. Croix, Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute, NIH, Frederick, Maryland 21702, United States

Martin J. Schnermann, Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States

REFERENCES

  • (1).Joshi BP; Wang TD Targeted Optical Imaging Agents in Cancer: Focus on Clinical Applications. Contrast Media Mol. Imaging 2018, 2018, 2015237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (2).Rosenthal EL; Warram JM; de Boer E; Chung TK; Korb ML; Brandwein-Gensler M; Strong TV; Schmalbach CE; Morlandt AB; Agarwal G; et al. Safety and Tumor Specificity of Cetuximab-IRDye800 for Surgical Navigation in Head and Neck Cancer. Clin Cancer Res 2015, 21, 3658–3666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (3).Chari RVJ; Miller ML; Widdison WC Antibody-drug conjugates: an emerging concept in cancer therapy. Am. Ethnol. 2014, 53, 3796–3827. [DOI] [PubMed] [Google Scholar]
  • (4).Usama SM; Thapaliya ER; Luciano MP; Schnermann MJ Not so innocent: Impact of fluorophore chemistry on the in vivo properties of bioconjugates. Curr. Opin. Chem. Biol. 2021, 63, 38–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (5).Donaghy H Effects of antibody, drug and linker on the preclinical and clinical toxicities of antibody-drug conjugates. mAbs 2016, 8, 659–671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (6).Buecheler JW; Winzer M; Weber C; Gieseler H Alteration of Physicochemical Properties for Antibody-Drug Conjugates and Their Impact on Stability. J. Pharm. Sci. 2020, 109, 161–168. [DOI] [PubMed] [Google Scholar]
  • (7).Agarwal P; Bertozzi CR Site-specific antibody-drug conjugates: the nexus of bioorthogonal chemistry, protein engineering, and drug development. Bioconjugate Chem. 2015, 26, 176–192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (8).Panowski S; Bhakta S; Raab H; Polakis P; Junutula JR Site-specific antibody drug conjugates for cancer therapy. mAbs 2014, 6, 34–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (9).Sochaj AM; Swiderska KW; Otlewski J Current methods for the synthesis of homogeneous antibody-drug conjugates. Biotechnol. Adv. 2015, 33, 775–784. [DOI] [PubMed] [Google Scholar]
  • (10).Ernst LA; Gupta RK; Mujumdar RB; Waggoner AS Cyanine dye labeling reagents for sulfhydryl groups. Cytometry 1989, 10, 3–10. [DOI] [PubMed] [Google Scholar]
  • (11).Nani RR; Shaum JB; Gorka AP; Schnermann MJ Electrophile-Integrating Smiles Rearrangement Provides Previously Inaccessible C4′-O-Alkyl Heptamethine Cyanine Fluorophores. Org. Lett. 2015, 17, 302–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (12).Sato K; Nagaya T; Nakamura Y; Harada T; Nani RR; Shaum JB; Gorka AP; Kim I; Paik CH; Choyke PL; Schnermann MJ; Kobayashi H Impact of C4′-O-Alkyl Linker on in Vivo Pharmacokinetics of Near-Infrared Cyanine/Monoclonal Antibody Conjugates. Mol. Pharmaceutics 2015, 12, 3303–3311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (13).Sato K; Gorka AP; Nagaya T; Michie MS; Nakamura Y; Nani RR; Coble VL; Vasalatiy OV; Swenson RE; Choyke PL; et al. Effect of charge localization on the: In vivo optical imaging properties of near-infrared cyanine dye/monoclonal antibody conjugates. Mol. BioSyst. 2016, 12, 3046–3056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (14).Sato K; Gorka AP; Nagaya T; Michie MS; Nani RR; Nakamura Y; Coble VL; Vasalatiy OV; Swenson RE; Choyke PL; Schnermann MJ; Kobayashi H Role of Fluorophore Charge on the in Vivo Optical Imaging Properties of Near-Infrared Cyanine Dye/Monoclonal Antibody Conjugates. Bioconjugate Chem. 2016, 27, 404–413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (15).Cha J; Nani RR; Luciano MP; Kline G; Broch A; Kim K; Namgoong JM; Kulkarni RA; Meier JL; et al. A chemically stable fluorescent marker of the ureter. Bioorg. Med. Chem. Lett. 2018, 28, 2741–2745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (16).Luciano MP; Crooke SN; Nourian S; Dingle I; Nani RR; Kline G; Patel NL; Robinson CM; Difilippantonio S; et al. A Nonaggregating Heptamethine Cyanine for Building Brighter Labeled Biomolecules. ACS Chem. Biol. 2019, 14, 934–940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (17).Li D-H; Schreiber CL; Smith BD Sterically Shielded Heptamethine Cyanine Dyes for Bioconjugation and High Performance Near-Infrared Fluorescence Imaging. Angewandte Chemie International Edition 2020, 59, 12154–12161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (18).Yazaki P; Lwin T; Minnix M; Li L; Sherman A; Molnar J; Miller A; Frankel P; Chea J; Poku E; Bowles N; Hoffman RM; Shively JE; Bouvet M Improved antibody-guided surgery with a near-infrared dye on a PEGylated linker for CEA-positive tumors. Journal of Biomedical Optics 2019, 24, No. 066012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (19).Rodell CB; Baldwin P; Fernandez B; Weissleder R; Sridhar S; Dubach JM Quantification of Cellular Drug Biodistribution Addresses Challenges in Evaluating in vitro and in vivo Encapsulated Drug Delivery. Adv Ther (Weinh) 2021, 4, 2000125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (20).Thurber GM; Yang KS; Reiner T; Kohler RH; Sorger P; Mitchison T; Weissleder R Single-cell and subcellular pharmacokinetic imaging allows insight into drug action in vivo. Nat. Commun. 2013, 4, 1504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (21).Cilliers C; Menezes B; Nessler I; Linderman J; Thurber GM Improved Tumor Penetration and Single-Cell Targeting of Antibody-Drug Conjugates Increases Anticancer Efficacy and Host Survival. Cancer Res. 2018, 78, 758–768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (22).Simmons JK; Burke PJ; Cochran JH; Pittman PG; Lyon RP Reducing the antigen-independent toxicity of antibody-drug conjugates by minimizing their non-specific clearance through PEGylation. Toxicol. Appl. Pharmacol. 2020, 392, 114932. [DOI] [PubMed] [Google Scholar]
  • (23).Lyon RP; Bovee TD; Doronina SO; Burke PJ; Hunter JH; Neff-LaFord HD; Jonas M; Anderson ME; Setter JR; Senter PD Reducing hydrophobicity of homogeneous antibody-drug conjugates improves pharmacokinetics and therapeutic index. Nat. Biotechnol. 2015, 33, 733–735. [DOI] [PubMed] [Google Scholar]
  • (24).Zhao RY; Wilhelm SD; Audette C; Jones G; Leece BA; Lazar AC; Goldmacher VS; Singh R; Kovtun Y; et al. Synthesis and evaluation of hydrophilic linkers for antibody-maytansinoid conjugates. J. Med. Chem. 2011, 54, 3606–3623. [DOI] [PubMed] [Google Scholar]
  • (25).Moon SJ; Govindan SV; Cardillo TM; D’Souza CA; Hansen HJ; Goldenberg DM Antibody conjugates of 7-ethyl-10-hydroxycamptothecin (SN-38) for targeted cancer chemotherapy. J. Med. Chem. 2008, 51, 6916–6926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (26).Miller ML; Roller EE; Zhao RY; Leece BA; Ab O; Baloglu E; Goldmacher VS; Chari RV Synthesis of taxoids with improved cytotoxicity and solubility for use in tumor-specific delivery. J. Med. Chem. 2004, 47, 4802–4805. [DOI] [PubMed] [Google Scholar]
  • (27).King HD; Dubowchik GM; Mastalerz H; Willner D; Hofstead SJ; Firestone RA; Lasch SJ; Trail PA Monoclonal antibody conjugates of doxorubicin prepared with branched peptide linkers: inhibition of aggregation by methoxytriethyleneglycol chains. J. Med. Chem. 2002, 45, 4336–4343. [DOI] [PubMed] [Google Scholar]
  • (28).Gruber HJ; Hahn CD; Kada G; Riener CK; Harms GS; Ahrer W; Dax TG; Knaus HG Anomalous fluorescence enhancement of Cy3 and cy3.5 versus anomalous fluorescence loss of Cy5 and Cy7 upon covalent linking to IgG and noncovalent binding to avidin. Bioconjugate Chem. 2000, 11, 696–704. [DOI] [PubMed] [Google Scholar]
  • (29).Pauli J; Grabolle M; Brehm R; Spieles M; Hamann FM; Wenzel M; Hilger I; Resch-Genger U Suitable labels for molecular imaging–influence of dye structure and hydrophilicity on the spectroscopic properties of IgG conjugates. Bioconjugate Chem. 2011, 22, 1298–1308. [DOI] [PubMed] [Google Scholar]
  • (30).Pauli J; Pochstein M; Haase A; Napp J; Luch A; Resch-Genger U Influence of Label and Charge Density on the Association of the Therapeutic Monoclonal Antibodies Trastuzumab and Cetuximab Conjugated to Anionic Fluorophores. ChemBioChem 2017, 18, 101–110. [DOI] [PubMed] [Google Scholar]
  • (31).Rijpkema M; Bos DL; Cornelissen AS; Franssen GM; Goldenberg DM; Oyen WJ; Boerman OC Optimization of Dual-Labeled Antibodies for Targeted Intraoperative Imaging of Tumors. Mol Imaging 2015, 14, 348–355. [PubMed] [Google Scholar]
  • (32).Cohen R; Stammes MA; de Roos IH; Stigter-van Walsum M; Visser GW; van Dongen GA Inert coupling of IRDye800CW to monoclonal antibodies for clinical optical imaging of tumor targets. EJNMMI Res 2011, 1, 31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (33).Conner KP; Rock BM; Kwon GK; Balthasar JP; Abuqayyas L; Wienkers LC; Rock DA Evaluation of near infrared fluorescent labeling of monoclonal antibodies as a tool for tissue distribution. Drug Metab. Dispos. 2014, 42, 1906–1913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (34).Burke PJ; Hamilton JZ; Jeffrey SC; Hunter JH; Doronina SO; Okeley NM; Miyamoto JB; Anderson ME; Stone IJ; Ulrich ML; Simmons JK; McKinney EE; Senter PD; Lyon RP Optimization of a PEGylated Glucuronide-Monomethylauristatin E Linker for Antibody-Drug Conjugates. Mol. Cancer Ther. 2017, 16, 116–123. [DOI] [PubMed] [Google Scholar]
  • (35).Mujumdar RB; Ernst LA; Mujumdar SR; Lewis CJ; Waggoner AS Cyanine Dye Labeling Reagents - Sulfoindocyanine Succinimidyl Esters. Bioconjugate Chem. 1993, 4, 105–111. [DOI] [PubMed] [Google Scholar]
  • (36).Mujumdar SR; Mujumdar RB; Grant CM; Waggoner AS Cyanine-labeling reagents: Sulfobenzindocyanine succinimidyl esters. Bioconjugate Chem. 1996, 7, 356–362. [DOI] [PubMed] [Google Scholar]
  • (37).Longmire M; Choyke PL; Kobayashi H Clearance properties of nano-sized particles and molecules as imaging agents: considerations and caveats. Nanomedicine (Lond) 2008, 3, 703–717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (38).Meyer DW; Bou LB; Shum S; Jonas M; Anderson ME; Hamilton JZ; Hunter JH; Wo SW; Wong AO; et al. An in Vitro Assay Using Cultured Kupffer Cells Can Predict the Impact of Drug Conjugation on in Vivo Antibody Pharmacokinetics. Mol. Pharmaceutics 2020, 17, 802–809. [DOI] [PubMed] [Google Scholar]
  • (39).Seaman S; Zhu Z; Saha S; Zhang XM; Yang MY; Hilton MB; Morris K; Szot C; Morris H; Swing DA; et al. Eradication of Tumors through Simultaneous Ablation of CD276/B7-H3-Positive Tumor Cells and Tumor Vasculature. Cancer Cell 2017, 31, 501–515.e8. e8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (40).Kendsersky NM; Lindsay J; Kolb EA; Smith MA; Teicher BA; Erickson SW; Earley EJ; Mosse YP; Martinez D; et al. The B7-H3-Targeting Antibody-Drug Conjugate m276-SL-PBD Is Potently Effective Against Pediatric Cancer Preclinical Solid Tumor Models. Clin Cancer Res 2021, 27, 2938–2946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (41).Qin X; Zhang H; Ye D; Dai B; Zhu Y; Shi G B7-H3 is a new cancer-specific endothelial marker in clear cell renal cell carcinoma. Onco Targets Ther 2013, 6, 1667–1673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (42).Zang X; Sullivan PS; Soslow RA; Waitz R; Reuter VE; Wilton A; Thaler HT; Arul M; Slovin SF; Wei J; et al. Tumor associated endothelial expression of B7-H3 predicts survival in ovarian carcinomas. Mod Pathol 2010, 23, 1104–1112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (43).Brunner A; Hinterholzer S; Riss P; Heinze G; Brustmann H Immunoexpression of B7-H3 in endometrial cancer: relation to tumor T-cell infiltration and prognosis. Gynecol Oncol 2012, 124, 105–111. [DOI] [PubMed] [Google Scholar]
  • (44).Seaman S; Stevens J; Yang MY; Logsdon D; Graff-Cherry C; St Croix B Genes that distinguish physiological and pathological angiogenesis. Cancer Cell 2007, 11, 539–554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (45).Choi HS; Nasr K; Alyabyev S; Feith D; Lee JH; Kim SH; Ashitate Y; Hyun H; Patonay G; Strekowski L; et al. Synthesis and In Vivo Fate of Zwitterionic Near-Infrared Fluorophores. Angew Chem Int Edit 2011, 50, 6258–6263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (46).Boswell CA; Tesar DB; Mukhyala K; Theil FP; Fielder PJ; Khawli LA Effects of charge on antibody tissue distribution and pharmacokinetics. Bioconjugate Chem. 2010, 21, 2153–2163. [DOI] [PubMed] [Google Scholar]
  • (47).Lee HJ; Pardridge WM Monoclonal antibody radiopharmaceuticals: cationization, pegylation, radiometal chelation, pharmacokinetics, and tumor imaging. Bioconjugate Chem. 2003, 14, 546–553. [DOI] [PubMed] [Google Scholar]
  • (48).Lee W; Bobba KN; Kim JY; Park H; Bhise A; Kim W; Lee K; Rajkumar S; Nam B; Lee KC; Lee SH; Ko S; Lee HJ; Jung ST; Yoo J A short PEG linker alters the in vivo pharmacokinetics of trastuzumab to yield high-contrast immuno-PET images. J. Mater. Chem. B 2021, 9, 2993–2997. [DOI] [PubMed] [Google Scholar]
  • (49).Guillou A; Earley DF; Klingler S; Nisli E; Nuesch LJ; Fay R; Holland JP The Influence of a Polyethylene Glycol Linker on the Metabolism and Pharmacokinetics of a (89)Zr-Radiolabeled Antibody. Bioconjugate Chem. 2021, 32, 1263–1275. [DOI] [PubMed] [Google Scholar]
  • (50).Sano K; Nakajima T; Miyazaki K; Ohuchi Y; Ikegami T; Choyke PL; Kobayashi H Short PEG-linkers improve the performance of targeted, activatable monoclonal antibody-indocyanine green optical imaging probes. Bioconjugate Chem. 2013, 24, 811–816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (51).Chapman AP PEGylated antibodies and antibody fragments for improved therapy: a review. Adv. Drug Delivery Rev. 2002, 54, 531–545. [DOI] [PubMed] [Google Scholar]
  • (52).Gokarn YR; McLean M; Laue TM Effect of PEGylation on protein hydrodynamics. Mol. Pharmaceutics 2012, 9, 762–773. [DOI] [PubMed] [Google Scholar]
  • (53).Takashina K; Kitamura K; Yamaguchi T; Noguchi A; Noguchi A; Tsurumi H; Takahashi T Comparative pharmacokinetic properties of murine monoclonal antibody A7 modified with neocarzinostatin, dextran and polyethylene glycol. Jpn. J. Cancer Res. 1991, 82, 1145–1150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (54).Kitamura K; Takahashi T; Yamaguchi T; Noguchi A; Noguchi A; Takashina K; Tsurumi H; Inagake M; Toyokuni T; Hakomori S Chemical engineering of the monoclonal antibody A7 by polyethylene glycol for targeting cancer chemotherapy. Cancer Res. 1991, 51, 4310–4315. [PubMed] [Google Scholar]
  • (55).Suzuki T; Kanbara N; Tomono T; Hayashi N; Shinohara I Physicochemical and biological properties of poly(ethylene glycol)-coupled immunoglobulin G. Biochim. Biophys. Acta 1984, 788, 248–255. [DOI] [PubMed] [Google Scholar]
  • (56).Elsadek NE; Hondo E; Shimizu T; Takata H; Abu Lila AS; Emam SE; Ando H; Ishima Y; Ishida T Impact of Pre-Existing or Induced Anti-PEG IgM on the Pharmacokinetics of Peginterferon Alfa-2a (Pegasys) in Mice. Mol. Pharmaceutics 2020, 17, 2964–2970. [DOI] [PubMed] [Google Scholar]
  • (57).Yang Q; Lai SK Anti-PEG immunity: emergence, characteristics, and unaddressed questions. Wiley Interdiscip Rev Nanomed Nanobiotechnol 2015, 7, 655–677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (58).Huckaby JT; Jacobs TM; Li Z; Robert JP; Wang A; Nicely NI; Lai SK Structure of an anti-PEG antibody reveals an open ring that captures highly flexible PEG polymers. Communications Chemistry 2020, 124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (59).Schlenoff JB Zwitteration: coating surfaces with zwitterionic functionality to reduce nonspecific adsorption. Langmuir 2014, 30, 9625–9636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (60).Erfani A; Seaberg J; Aichele CP; Ramsey JD Interactions between Biomolecules and Zwitterionic Moieties: A Review. Biomacromolecules 2020, 21, 2557–2573. [DOI] [PubMed] [Google Scholar]
  • (61).Debie P; Van Quathem J; Hansen I; Bala G; Massa S; Devoogdt N; Xavier C; Hernot S Effect of Dye and Conjugation Chemistry on the Biodistribution Profile of Near-Infrared-Labeled Nanobodies as Tracers for Image-Guided Surgery. Mol. Pharmaceutics 2017, 14, 1145–1153. [DOI] [PubMed] [Google Scholar]
  • (62).Luciano MP; Dingle I; Nourian S; Schnermann MJ Preferential Light-Chain Labeling of Native Monoclonal Antibodies Improves the Properties of Fluorophore Conjugates. Tetrahedron Lett. 2021, 75, 153211. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

35389618_Schnermann_SM

RESOURCES