Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Jul 6.
Published in final edited form as: Tetrahedron Lett. 2021 May 27;75:153211. doi: 10.1016/j.tetlet.2021.153211

Preferential Light-Chain Labeling of Native Monoclonal Antibodies Improves the Properties of Fluorophore Conjugates

Michael P Luciano 1, Ivan Dingle 1, Saghar Nourian 1, Martin J Schnermann 1
PMCID: PMC8312683  NIHMSID: NIHMS1714572  PMID: 34321699

Abstract

Site specific labeling methods have significant potential to enhance the properties of antibody conjugates. While studied extensively in the context of antibody-drug conjugates (ADCs), few studies have examined the impact of homogenous labeling on the properties of antibody-fluorophore conjugates (AFCs). We report the application of pentafluorophenyl (PFP) esters, which had previously been shown to be reasonably selective for K188 of the kappa light chain of human IGG antibodies, toward producing AFCs. We show that simple replacement of N-hydroxy succinimide (NHS) with PFP dramatically increases the light-chain specificity of near-infrared (NIR) AFCs. Comparing the properties of AFCs labeled using NHS and PFP-activated esters reveals that the latter exhibits reduced aggregation and improved brightness, both in vitro and in vivo. Overall, the use of PFP esters provides a remarkably simple approach to provide selectively labeled antibodies with improved properties.

Graphical Abstract

graphic file with name nihms-1714572-f0001.jpg

Introduction

Antibody-drug conjugates (ADCs) are an emerging approach for cancer therapy that combine the potency of small molecule anticancer drugs with the ability to target tumor-associated antigens.13 Related antibody-fluorophore conjugates (AFCs) enable the optical detection of target-expressing cells, with clinical applications in the context of fluorescence-guided surgery (FGS).4, 5 Numerous AFC constructs are currently being clinically evaluated for FGS, with most consisting of clinically approved therapeutic monoclonal antibodies (mAbs) and the heptamethine cyanine IR-800CW.5 By far the most common way to prepare ADCs and AFCs is through amide formation between a lysine residue on the antibody and an active ester (i.e. NHS ester) of the payload.611 Due to the abundance of lysine residues (>50 in a typical IgG1) and the high reactivity of NHS esters, heterogeneous mixtures are obtained. For ADCs, this heterogeneity is, at a minimum, an issue for batch-to-batch reproducibility and, at worst, can reduce the therapeutic index.12 For AFCs, the impact of homogenous labeling has not been studied extensively, though deleterious effects of over-labeled conjugates have been proposed.13

To circumvent these issues, a number of approaches have been developed to produce increasingly homogeneous small molecule-antibody conjugates through site-specific labeling chemistry.2, 1416 The most well-studied is the Thiomab approach developed by Genentech, in which a cysteine residue is installed on the protein and labeled using maleimide or related thiol-reactive chemistry. In early studies, this approach was shown to improve the therapeutic index of an anti-MUC16-MMAE ADC.17 Depending on the site in which the engineered cysteine was introduced, this approach can improve the in vivo pharmacokinetics and metabolic stability of ADCs.18, 19 On the other hand, the effect of labeling chemistry on the properties of AFCs is much less studied, and we are only aware of three prior examples of homogenously-produced protein-NIR fluorophore conjugates.2022

Of particular interest are site-selective labeling methods that can be used on native mAbs. Such approaches benefit from employing broadly available fully humanized mAbs, which have been the basis of numerous FDA-approved therapuetics.23 A unique approach to site-specific labeling of native mAbs is the observation that a single lysine residue (K188) in the kappa (κ) light chain constant domain of human mAbs could be preferentially labeled (w/ ~70% selectivity) by employing pentafluorophenyl (PFP) esters as the active ester species for the conjugation.24 Building on this work in the patent literature, Pham and co-workers studied the mechanism of this selectivity in greater detail. They found that highly homogeneous (>95% selectivity) Fab fragment conjugates can be obtained with preferential K188 labeling by optimization of the reaction conditions (4 °C or flow chemistry) or in combination with specific point mutations in the κ light chain region.25 This preferential light-chain labeling strategy, although demonstrating only ~70% selectivity for native intact mAbs, represented a straightforward way for us to study the effect of labeling chemistry/site on the properties of AFCs.

Herein we report our initial findings applying this labeling strategy to a heptamethine cyanine fluorophore. Labeling the anti-EGFR mAb Panitumumab with an NHS ester leads to AFCs that are preferentially labeled on the heavy chain and exhibit significant fluorescence quenching. On the other hand, using a PFP ester provides conjugates with preferential light chain labeling. These selectively labeled bioconjugates are significantly brighter and exhibit higher tumor signals when administered intravenously to tumor-bearing mice. Overall, this approach, which is notably straightforward, demonstrates that labeling chemistry can significantly impact the properties of AFCs.

Our studies focused on the model C4’-O-alkyl heptamethine cyanine fluorophore diSulfo-FNIR, which we prepared from C4’-amine-substituted 1 using our previously reported electrophile-integrating Smiles rearrangement chemistry (Scheme 1).26 This chemistry enables the facile preparation of C4’-O-alkyl heptamethine cyanines, which have improved stability to the presence of biological thiols compared to C4’-O-aryl substituted derivatives.26, 27 We have used this chemistry over the last several years to study the effect of substituents and charge balance on the properties of heptamethine cyanines in the context of AFCs, which culminated in the development of the efficient mAb label, FNIR-Tag.2831 In practice, diSulfo-FNIR could be obtained from 1 and glutaric anhydride, as we previously reported (Scheme 1).32 DiSulfo-FNIR could be converted to the corresponding NHS-ester, diSulfo-FNIR-NHS using TSTU. Alternatively, the corresponding PFP ester, diSulfo-FNIR-PFP, could be accessed using the commercially available bis(pentafluorophenyl) carbonate and N-methylmorpholine.

Scheme 1.

Scheme 1.

Synthesis of N-hydroxysuccinimdyl (NHS) and pentafluorophenyl (PFP) reactive esters of diSulfo-FNIR.

We then examined a variety of labeling conditions using diSulfo-FNIR-PFP. Our initial screening conditions used Panitumumab at a concentration of 7 μM in different buffers (PBS or HEPES), with a 10-fold molar excess of diSulfo-FNIR-PFP at 4 °C for 18 h (Table 1). We experimented with the addition of an organic cosolvent (up to 20%). Through this screening, we found that using pH 7.0 PBS rather than typically employed pH 8.0 PBS buffer produced conjugates with improved light chain to heavy chain (LC:HC) ratios as determined by reducing SDS-PAGE. We also found PBS with 10% DMF was preferable to HEPES buffer. Although these conditions produced a favorable LC:HC ratio, the degree of labeling (DOL) was lower than expected (~ 1.1:1). Therefore, we examined sequential addition of the active pentafluorophenyl ester (2x) separated by 3 hours, which improved the DOL considerably to a value of 1.7, while retaining the same LC:HC selectivity (1.7:1). This conjugate, PFP-FNIR-Pan, was prepared on up to a 2 mg scale and was purified using centrifugal filtration followed by dialysis. Of note, to date, mass spectral analysis of the conjugates modified with this charged fluorophore has been quite difficult, and we have not been able to analyze them to a finer level of molecular detail. However, we hypothesize that the selective light chain labeling we observe likely reflects at least preferential formation of K188 conjugates, as reported previously.25

Table 1.

Reaction conditions screening for selective light chain labeling.

Entry R Buffer/Co-Solvent Conc. T Time LC:HC
1 PFP 40 mM PBS (pH 8)/20% DMF 7 μM 4 °C 16 h 0.7
2 PFP 40 mM PBS (pH 8)/5% DMF 7 μM 4 °C 16 h 0.8
3 PFP 40 mM HEPES (pH 8)/10% DMF 7 μM 4 °C 16 h 0.7
4 PFP 40 mM HEPES (pH 7)/10% DMF 7 μM 4 °C 16 h 1.1
5 PFP 40 mM PBS (pH 8)/10% DMF 7 μM 4 °C 16 h 0.8
6 PFP 40 mM PBS (pH 7)/10% DMF 7 μM 4 °C 16 h 1.7
7 PFP 40 mM HEPES (pH 8)/1% DMF 7 μM 4 °C 16 h 0.5
8 PFP 40 mM HEPES (pH 7)/1% DMF 7 μM 4 °C 16 h 0.8
9 PFP 40 mM PBS (pH 8)/1% DMF 7 μM 4 °C 16 h 0.6
10 PFP 40 mM PBS (pH 7)/1% DMF 7 μM 4 °C 16 h 0.7
11 NHS 1M PBS (pH 8.5) 500 μM r.t. 1 h 0.1

In order to evaluate the properties of labeled PFP-FNIR-Pan, we prepared the corresponding conjugate with diSulfo-FNIR-NHS, NHS-FNIR-Pan, with a matching DOL of 1.7. In this case, the LC:HC ratio is 0.1 (Fig. 2) – highlighting the dramatic effect of the PFP labeling strategy. Of note, using the optimized conditions for PFP-FNIR-Pan with diSulfo-FNIR-NHS, resulted in only a modest improvement in the LC:HC selectivity from 0.1 to 0.2 (Fig. S1). This result demonstrates that the PFP ester, not the labeling conditions, are responsible for the enhanced light chain selectivity.

Figure 2.

Figure 2.

a) General scheme for labeling conditions screening using the model fluorophore diSulfo-FNIR. b) Gel electrophoresis (Cy7 excitation) of diSulfo-FNIR labeled light and heavy chain fragments after SDS-PAGE (reducing conditions). The light chain: heavy chain ratio (LC:HC) was determined by dividing the quantified signals from the two fragments.

The absorbance and emission spectra of the two conjugates were obtained in pH 7.4 PBS (50 mM) at a 500 nM mAb concentration (Fig. 3A). As expected, with NHS-FNIR-Pan the absorbance spectra contained a blue-shifted band arising from the nonemissive H-aggregate dimer species.33, 34 By contrast, the absorbance spectrum of PFP-FNIR-Pan more closely resembles that of the free dye (Fig S2). The dramatic difference between the optical properties of the conjugates is also readily observed by the difference in emission intensity. When solutions with equal mAb concentration (500 nM) were excited at 730 nm, PFP-FNIR-Pan was ~3x brighter than the NHS-FNIR-Pan conjugate. In line with previous observations, dilution of the same conjugates into MeOH:PBS (protein denaturing conditions) restores the emission intensity of the non-selectively labeled control, albeit not completely (Fig. S3). We then examined the two conjugates by flow cytometry to determine the impact of labeling method on cellular uptake. When incubating PFP-FNIR-Pan or NHS-FNIR-Pan with MDA-MB-468 cells (EGFR+), we observed the PFP-labeled mAbs was significantly brighter as measured by the mean fluorescence intensity of the labeled cells (Fig. 3B). By contrast, no significant uptake of either of the conjugates was observed in MCF-7 cells (EGFR–).

Figure 3.

Figure 3.

a) Absorbance and emission spectra of NHS-FNIR-Pan, black trace and PFP-FNIR-Pan, red trace with a DOL of 1.7 and a mAb concentration of 500 nM in PBS (50 mM. pH 7.4). b) Flow cytometric analysis of DOL 1.7 PFP-FNIR-Pan and PFP-FNIR-Pan in MCF-7 (EGFR –) and MDA-MB-468 (EGFR+) cells.

Finally, we examined the difference between these two conjugates in the context of in vivo tumor imaging. Prior studies with AFCs have demonstrated that even moderate changes to the fluorophore label density has an impact on the clearance of AFCs.29 We have also found that AFC aggregation (due to high label density) and poor pharmacokinetics are related, often resulting in increased liver uptake.28,33 Equal doses of NHS-FNIR-Pan and PFP-FNIR-Pan (10 ug, DOL 1.7) were administered intravenously in MDA-MB-468 xenograft (n = 5 per group) bearing mice. The PFP-FNIR-Pan exhibited significantly improved tumor uptake relative to NHS-FNIR-Pan at all timepoints and significantly higher tumor to background ratios (TBR) at 48 h and 1 week post injection (Fig. 4).

Figure 4.

Figure 4.

a) Representative side and ventral view images of MDA-MB-468 xenograft tumor-bearing mice (n = 5) that received either NHS-FNIR-Pan or PFP-FNIR-Pan, mice were dosed by tail vein injection (10 ug Ab, DOL 1.7, 1.2 nmol effective dye dose). b) Quantification of Tumor Signal and Tumor to background ratios (TBR) of the two conjugates at the timepoints indicated. The tumor signal values, as determined by a region of interest (ROI) were normalized to tumor size for each mouse. Data points are displayed as mean ± SD, and the p-values were evaluated by student t-test (*** p-value ≤ 0.001).

In this study, we have employed a selective mAb labeling technique to produce AFCs with enriched labeling in the LC constant domain of a human mAb (Panitumumab). The selectivity of the LC chain labeling observed with a charged fluorophore was similar to early reports that mainly employed hydrophobic, uncharged payloads.25 Notably, the selectively labeled conjugates showed no signs of aggregation in their optical spectra, retained fluorescence emission similar to that of the unconjugated dye, and showed improved uptake in MDA-MB-468 cells relative to the random lysine-labeled conjugate using diSulfo-FNIR-NHS. Additionally, PFP-FNIR-Pan exhibited significantly higher tumor signal than NHS-FNIR-Pan in an MDA-MB-468 xenograft mouse model.

We hope these studies highlight the potential for this simple PFP-based method to provide antibody conjugates of improved homogeneity. Additionally, this work provides further insight into the relationship between fluorophore labeling site and AFC properties. These studies indicate more homogeneous conjugates reduce the propensity for fluorophore H-aggregates to form. This property, in turn, leads to improved optical properties and tumor signal. In combination with prior work, we and others have now shown that the properties of AFCs can be improved by either homogenous labeling or by the use of highly polar, but net-neutral (i.e. zwitterionic), fluorophores.28,33 Notably, however, the latter benefits from achieving higher DOL. Going forward, we anticipate that, much as the properties of ADCs have been improved through the use homogenous protein labeling methods, the same will be true for AFCs.

Supplementary Material

1

Figure 1.

Figure 1.

a) Structure of model heptamethine cyanine fluorophore diSulfo-FNIR and the activated esters used in this study. b) General schematic for Panitumumab labeling with diSulfo-FNIR activated esters to produce either random lysine labeled (NHS) or selective light chain labeling (PFP).

Highlights.

Pentafluorophenyl esters of cyanine fluorophores can be readily prepared

These esters selectively label the light chain of native monoclonal antibodies

The resulting conjugates are brighter in vitro and in vivo

Acknowledgements

We thank Dr. Joseph Barchi, NCI Center for Cancer Research (NCI-CCR), for NMR assistance and Dr. James Kelley, NCI-CCR, for mass spectrometric analysis. We thank Drs. Nimit L. Patel, Lisa Riffle and Joseph D. Kalen (Small Animal Imaging Program) and Chelsea Sanders and Simone Difilippantonio (Laboratory Animal Sciences Program) for assistance with the in vivo study. This work was supported by the Intramural Research Program of the National Institutes of Health (NIH), NCI-CCR.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

This paper is dedicated to Prof. Dale L. Boger on the occasion of his receipt of the 2020 Tetrahedron Prize for Creativity.

Animal Care and Use

In vivo studies were performed according to the Frederick National Laboratory for Cancer Research (Frederick, MD) Animal Care and Use committee guidelines. Frederick National Laboratory is accredited by AAALAC International and follows the Public Health Service Policy for the Care and Use of Laboratory Animals. Animal care was provided in accordance with the procedures outlined in the “Guide for Care and Use of Laboratory Animals” (National Research Council; 2011; National Academies Press; Washington, D.C.).

References

  • 1.Thomas A; Teicher BA; Hassan R The Lancet Oncology 2016, 17, e254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Beck A; Goetsch L; Dumontet C; Corvaia N Nat Rev Drug Discov 2017, 16, 315. [DOI] [PubMed] [Google Scholar]
  • 3.Cheever MA; Allison JP; Ferris AS; Finn OJ; Hastings BM; Hecht TT; Mellman I; Prindiville SA; Viner JL; Weiner LM; Matrisian LM Clin Cancer Res 2009, 15, 5323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Hernot S; van Manen L; Debie P; Mieog JSD; Vahrmeijer AL The Lancet Oncology 2019, 20, e354. [DOI] [PubMed] [Google Scholar]
  • 5.Barth CW; Gibbs SL Proc SPIE Int Soc Opt Eng 2020, 11222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Gutowski M; Framery B; Boonstra MC; Garambois V; Quenet F; Dumas K; Scherninski F; Cailler F; Vahrmeijer AL; Pelegrin A Surg Oncol 2017, 26, 153. [DOI] [PubMed] [Google Scholar]
  • 7.Heath CH; Deep NL; Sweeny L; Zinn KR; Rosenthal EL Ann Surg Oncol 2012, 19, 3879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Hekman MC; Rijpkema M; Muselaers CH; Oosterwijk E; Hulsbergen-Van de Kaa CA; Boerman OC; Oyen WJ; Langenhuijsen JF; Mulders PF Theranostics 2018, 8, 2161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Hekman MCH; Rijpkema M; Bos DL; Oosterwijk E; Goldenberg DM; Mulders PFA; Boerman OC J Nucl Med 2017, 58, 706. [DOI] [PubMed] [Google Scholar]
  • 10.Korb ML; Hartman YE; Kovar J; Zinn KR; Bland KI; Rosenthal EL J Surg Res 2014, 188, 119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Terwisscha van Scheltinga AG; van Dam GM; Nagengast WB; Ntziachristos V; Hollema H; Herek JL; Schroder CP; Kosterink JG; Lub-de Hoog MN; de Vries EG J Nucl Med 2011, 52, 1778. [DOI] [PubMed] [Google Scholar]
  • 12.Ricart AD Clin Cancer Res 2011, 17, 6417. [DOI] [PubMed] [Google Scholar]
  • 13.Usama SM; Thapaliya ER; Luciano MP; Schnermann MJ Curr Opin Chem Biol 2021, 63, 38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Agarwal P; Bertozzi CR Bioconjug Chem 2015, 26, 176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Panowski S; Bhakta S; Raab H; Polakis P; Junutula JR Mabs-Austin 2014, 6, 34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Massa S; Xavier C; Muyldermans S; Devoogdt N Expert Opin Drug Deliv 2016, 13, 1149. [DOI] [PubMed] [Google Scholar]
  • 17.Junutula JR; Raab H; Clark S; Bhakta S; Leipold DD; Weir S; Chen Y; Simpson M; Tsai SP; Dennis MS; Lu Y; Meng YG; Ng C; Yang J; Lee CC; Duenas E; Gorrell J; Katta V; Kim A; McDorman K; Flagella K; Venook R; Ross S; Spencer SD; Lee Wong W; Lowman HB; Vandlen R; Sliwkowski MX; Scheller RH; Polakis P; Mallet W Nat Biotechnol 2008, 26, 925. [DOI] [PubMed] [Google Scholar]
  • 18.Ohri R; Bhakta S; Fourie-O’Donohue A; Dela Cruz-Chuh J; Tsai SP; Cook R; Wei B; Ng C; Wong AW; Bos AB; Farahi F; Bhakta J; Pillow TH; Raab H; Vandlen R; Polakis P; Liu Y; Erickson H; Junutula JR; Kozak KR Bioconjug Chem 2018, 29, 473. [DOI] [PubMed] [Google Scholar]
  • 19.Su D; Kozak KR; Sadowsky J; Yu SF; Fourie-O’Donohue A; Nelson C; Vandlen R; Ohri R; Liu L; Ng C; He J; Davis H; Lau J; Del Rosario G; Cosino E; Cruz-Chuh JD; Ma Y; Zhang D; Darwish M; Cai W; Chen C; Zhou H; Lu J; Liu Y; Kaur S; Xu K; Pillow TH Bioconjug Chem 2018, 29, 1155. [DOI] [PubMed] [Google Scholar]
  • 20.Debie P; Van Quathem J; Hansen I; Bala G; Massa S; Devoogdt N; Xavier C; Hernot S Mol Pharm 2017, 14, 1145. [DOI] [PubMed] [Google Scholar]
  • 21.Fung K; Sharma SK; Keinanen O; Roche KL; Lewis JS; Zeglis BM Mol Pharm 2020, 17, 3140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Houghton JL; Zeglis BM; Abdel-Atti D; Aggeler R; Sawada R; Agnew BJ; Scholz WW; Lewis JS Proc Natl Acad Sci U S A 2015, 112, 15850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Singh S; Kumar NK; Dwiwedi P; Charan J; Kaur R; Sidhu P; Chugh VK Curr Clin Pharmacol 2018, 13, 85. [DOI] [PubMed] [Google Scholar]
  • 24.Bhat AS; Bradshaw CW; Laurent OA; Lee A; Preston RR; Tumelty D; Wood LD; Yu WH In Multifunctional Antibody Conjugates, 2014, US Patent # 8,741,291 B2.
  • 25.Pham GH; Ou W; Bursulaya B; DiDonato M; Herath A; Jin Y; Hao X; Loren J; Spraggon G; Brock A; Uno T; Geierstanger BH; Cellitti SE Chembiochem 2018, 19, 799. [DOI] [PubMed] [Google Scholar]
  • 26.Nani RR; Shaum JB; Gorka AP; Schnermann MJ Org Lett 2015, 17, 302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Cha J; Nani RR; Luciano MP; Kline G; Broch A; Kim K; Namgoong JM; Kulkarni RA; Meier JL; Kim P; Schnermann MJ Bioorg Med Chem Lett 2018, 28, 2741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Luciano MP; Crooke SN; Nourian S; Dingle I; Nani RR; Kline G; Patel NL; Robinson CM; Difilippantonio S; Kalen JD; Finn MG; Schnermann MJ Acs Chem Biol 2019, 14, 934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Sato K; Gorka AP; Nagaya T; Michie MS; Nakamura Y; Nani RR; Coble VL; Vasalatiy OV; Swenson RE; Choyke PL; Schnermann MJ; Kobayashi H Mol Biosyst 2016, 12, 3046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Sato K; Gorka AP; Nagaya T; Michie MS; Nani RR; Nakamura Y; Coble VL; Vasalatiy OV; Swenson RE; Choyke PL; Schnermann MJ; Kobayashi H Bioconjug Chem 2016, 27, 404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Sato K; Nagaya T; Nakamura Y; Harada T; Nani RR; Shaum JB; Gorka AP; Kim I; Paik CH; Choyke PL; Schnermann MJ; Kobayashi H Mol Pharm 2015, 12, 3303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Luciano MP; Namgoong JM; Nani RR; Nam SH; Lee C; Shin IH; Schnermann MJ; Cha J Mol Pharm 2019, 16, 3253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Li DH; Schreiber CL; Smith BD Angew Chem Int Ed Engl 2020, 59, 12154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Hyun H; Owens EA; Narayana L; Wada H; Gravier J; Bao K; Frangioni JV; Choi HS; Henary M Rsc Adv 2014, 4, 58762. [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

1

RESOURCES