Open Access Article
This Open Access Article is licensed under a
Creative Commons Attribution 3.0 Unported Licence

A toolset of functionalized porphyrins with different linker strategies for application in bioconjugation

M. H. Staegemann ab, S. Gräfe b, R. Haag *a and A. Wiehe *ab
aInstitut für Chemie und Biochemie, Freie Universität Berlin, Takustrasse 3, 14195 Berlin, Germany. E-mail: haag@chemie.fu-berlin.de
bBiolitec research GmbH, Otto-Schott-Strasse 15, 07745 Jena, Germany. E-mail: arno.wiehe@biolitec.com

Received 20th July 2016 , Accepted 29th August 2016

First published on 30th August 2016


Abstract

The reaction of amines with pentafluorophenyl-substituted A3B-porphyrins has been used to obtain different useful reactive groups for further functionalization and/or conjugation of these porphyrins to other substrates or materials. Porphyrins with alkenyl, alkynyl, amino, azido, epoxide, hydroxyl, and maleimido groups have thus been synthesized. For the first time such functionalized porphyrins have been conjugated to hyperbranched polyglycerol (hPG) as a biocompatible carrier system for photodynamic therapy (PDT) using the copper(I)-catalyzed 1,3-dipolar cycloaddition (CuAAC). The photocytotoxicity of selected porphyrins as well as of the porphyrin-hPG-conjugates has been assessed in cellular assays with human epidermoid carcinoma A-253 and squamous carcinoma CAL-27 cells. For several biomedical applications a release of the active drug and/or fluorescent dye is desired. Therefore, additionally, the synthesis of A3B-porphyrins with cleavable linker moieties is presented, namely disulfide, cleavable in a reductive environment, and acetal linkers whose cleavage is pH triggered.


Introduction

Cyclic tetrapyrrolic systems are essential in many biological processes and are also of interest for diverse applications such as photodynamic therapy (PDT),1–6 light-harvesting,7,8 or catalysis.9–11 PDT is a treatment modality for malignant tissues, which is today routinely applied for the treatment of certain forms of cancer.1–6 In PDT, a dye – the so-called photosensitizer – and light are combined to provoke a toxic effect in the tumor cells. Different photosensitizers based on tetrapyrrolic structures are described in literature: e.g. chlorins and bacteriochlorins,2,12–17 porphyrins,2,5,6 phthalocyanines,18,19 and corroles.20,21 When choosing porphyrins as tetrapyrrolic systems, these may also be transformed into the corresponding chlorins or bacteriochlorins which are even more potent photosensitizers.2,12–17 If the connection to carriers or other substrates is intended porphyrins of the A3B-type (with ‘B’ being the substituent suitable for coupling) are preferable to assure a specific linkage without undesired crosslinking. One way to obtain such specifically functionalized tetrapyrroles is the nucleophilic aromatic substitution reaction of a fluorine atom in pentafluorophenyl-substituted tetrapyrrolic systems. Different nucleophiles have been used like amines,5,22–25 alcohols,5,26–28 carborane,29 phosphanes,30 phosphite,31 and thiols.14,26,32 Thereby, the reaction with amines or thiols does not require any addition of catalysts or other reagents (e.g. bases),5,22–25 which simplifies reaction conduct and workup.

In this work the functionalization of A3B-type pentafluorophenyl containing porphyrins with amines is described, specifically intended for conjugation of these porphyrins to other substrates, carrier systems or material surfaces. An easy and convenient way is shown to introduce the following functionalities: alkenyl, alkynyl, amino, azido, epoxide, hydroxyl, and maleimido. The alkynyl-substituted porphyrin was chosen for further linkage – via the copper(I)-catalyzed 1,3-dipolar cycloaddition (CuAAC) – to a second porphyrin, to glyco-substituents, and especially to hyperbranched polyglycerol (hPG), as a prominent example for a biocompatible carrier system,33–37 exemplifying the applicability of this method. One of the important issues with respect to carrier systems for medically active substances is the site-specific release of the active substance from the carrier.38–41 To provide such cleavable linkages porphyrins carrying disulfide or acetal linkers are also presented.

Synthesis

The focus of this work is the synthesis of substituted porphyrin systems to obtain a toolkit for cleavable and non-cleavable linkers to different substrates e.g. carrier systems, surfaces or the formation of multimeric systems. In literature different tetrapyrroles have been described and used for further linkage.42–44 Only little has been reported in this respect on the synthesis of porphyrins with cleavable linkers that should allow the release from a substrate or carrier system, which is of interest for many biological applications.45–47

In the literature a number of nucleophilic aromatic substitutions with amines on pentafluorophenyl-substituted porphyrins have been described involving however mainly the tetra-substituted derivatives.5,22–26,30 For the purpose of specific linkage mono-pentafluorophenyl-substituted porphyrins (A3B systems) are preferable therefore we expanded the substitution reaction onto these porphyrin systems (Scheme 1).


image file: c6ob01551d-s1.tif
Scheme 1 Regioselective nucleophilic aromatic substitution of A3B porphyrins 1a–c with different amines. R2-NH2 is defined as in Table 1. Reagents and conditions: DMSO, 0.5–7 h, 83–100 °C (detailed conditions and yields are given in Table 1).

The reaction was performed with porphyrins carrying 3-acetoxyphenyl 1a,27,48 3-benzyloxyphenyl 1b[thin space (1/6-em)]27 or 3-hydroxyphenyl 1c[thin space (1/6-em)]27,48 groups as R1 (substituent A). The structure of the A3B porphyrins with (protected) 3-hydroxyphenyl groups is inspired by the structure of the photosensitizer Temoporfin (5,10,15,20-tetrakis(3-hydroxyphenyl)-chlorin, mTHPC) which is one of the few photosensitizers currently approved for clinical use.49 The polar hydroxyphenyl groups thereby increase the solubility of the hydrophobic macrocycle in the biological environment and enhance membrane affinity.49

The different amines and detailed conditions are given in Table 1. The reaction was performed in DMSO at 83 °C (b.p. of propargylamine) or 100 °C. The reaction with the diamine cystamine under these conditions led to degradation of the disulfide linker resulting in low yield (results not shown). Therefore the reaction was tried under microwave conditions (Table 1, entry 1). Using the microwave the reaction time gets shorter at the same time the yield is improved, showing that with this method it is possible to introduce labile functionalities, like the disulfide-containing cystamine. The different polarities of R1 did not interfere with the reactivity of the amines, therefore unpolar substituents like 3-benzyloxyphenyl can be used as well as the polar 3-hydroxyphenyl group. However, the more polar 3-hydroxyphenyl group is of higher interest for biological applications due to its close analogy to the photosensitizer Temoporfin.

Table 1 Reactions of the A3B porphyrins 1a–c with amines in DMSO
Entry Starting material Amine R1 Conditionsa Product Yieldb [%]
a All the reactions were carried under argon in a sealed reaction vessel. b Yield of isolated product after purification. c In product R1 = H; the basic propargylamine simultaneously removes the acetyl protection groups.
1 1c Cystamine H 30 min, 100 °C microwave (300W) 2a 87
2 1c 1,4-Diaminobutane H 1 h, 100 °C 2b 69
4 1c 1,5-Diaminopentane H 1 h, 100 °C 2c 54
5 1c 1,6-Diaminohexane H 1 h, 100 °C 2d 79
3 1c 1-(N-Boc-),5-diaminopentane H 4 h, 100 °C 2e 69
6 1a Propargylamine Ac 3 h, 83 °C 2f[thin space (1/6-em)]c 94
7 1b Propargylamine Bn 7 h, 100 °C 2g 78


Employing the 3-acetoxyphenyl residues it is possible to do a two-step one-pot reaction.27,48 The amine acts as a nucleophile for the nucleophilic aromatic substitution and simultaneously removes the acetoxy protection groups resulting in the functionalized A3B-porphyrin with three polar hydroxyphenyl groups. This is shown with the example of the acetoxy-protected porphyrin 1a which on reaction with excess propargylamine directly afforded the deprotected and propargylamino-substituted compound 2f. This simplifies the synthesis of substituted A3B porphyrins and makes it possible to get to the final product in only two steps starting from pyrrole and aldehydes. The unsubstituted and the two propargylamino-substituted porphyrins 1c and 2f,g (Scheme 2) were further converted to their corresponding zinc-complexes 1d and 2h,i obtained between 73% and quant. yield.


image file: c6ob01551d-s2.tif
Scheme 2 Zinc insertion into the A3B porphyrins 1c, 2f and 2g. Reagents and conditions: Zn(OAc)2, NaOAc, MeOH or MeOH/DCM, 1–2 h, RT.

Mono-functionalized porphyrins like 2a–g should in principle also be accessible by the mono-functionalization of the tetrakispentafluorophenyl-substituted porphyrin 3 (Scheme 3). To test this 3 was reacted with propargylamine. Under optimized reaction conditions (DMSO/THF mixture, 1.5 h reaction time) the mono-propargylamino-substituted porphyrin 4 could be obtained in 22% yield, in addition, the disubstituted compound carrying two propargylamino-substituents was also isolated (18%, not shown). The A3B porphyrin 4 carries only one propargylamino-substituent it lacks, however, the polar hydroxyphenyl-substitution of 2a–g which significantly contributes to the solubility of the hydrophobic macrocycle in the biological environment.49 To overcome this, a subsequent modification of the three remaining pentafluoropheny-substituents e.g. by nucleophilic substitution would be necessary.


image file: c6ob01551d-s3.tif
Scheme 3 Synthesis of the mono-functionalized porphyrin 4. Reagents and conditions: propargylamine, DMSO/THF (1/1), 1.5 h, 100 °C.

The free amino group of the porphyrins 2a–d is a useful and reactive functionality for further modifications. It is possible to use it directly for the linkage to carriers or substrates. Use of amide coupling, e.g. allows the introduction of other linkage functionalities (Scheme 4). On the one hand it is possible to directly use a carboxylic acid, here propynoic acid, with DCC and 1-hydroxybenzotriazole hydrate (HOBt hydrate). This method is commonly used in peptide synthesis and prevents the formation of N-acylurea.50 The porphyrins 2a and 2b, propynoic acid, HOBt hydrate, and dicyclohexylcarbodiimide (DCC) in THF were stirred for 130 min at RT. The crude products were purified by column chromatography to afford the porphyrins 5a and 5b with a yield of 33 and 77%, respectively. Products 5a,b and the zinc complex 5c carry the alkyne functionality which allows the CuAAC in further reactions; in addition 5b and 5c incorporate a cleavable disulfide linker as well.


image file: c6ob01551d-s4.tif
Scheme 4 Substitution of the A3B porphyrins 2a–d with free amine end groups via amide coupling with propynoic acid or 3-(maleimido)propionic acid N-hydroxysuccinimide ester. Reagents and conditions: (i) propynoic acid, HOBt hydrate, DCC, THF, 130 min, RT; (ii) 3-(maleimido)propionic acid N-hydroxysuccinimide, DMF, 1 h., RT; (iii) Zn(OAc)2, NaOAc, MeOH, 30 min, RT (see Experimental section for further details).

On the other hand we used an active ester, which allows reactions with compounds containing amino-sensitive groups. One example is the maleimido functionality, which can undergo a reaction with the free amine of the porphyrin. Scheme 4 shows the reaction of 3-(maleimido)propionic acid N-hydroxysuccinimide ester with the porphyrins 2a, 2c and 2d.

The porphyrins 2a,c,d and 3-(maleimido)propionic acid N-hydroxysuccinimide ester were stirred in DMF for 1 h at RT. The crude products were purified by column chromatography to afford the porphyrins 6a–c in high yields between 65 and 81%. The introduced maleimido functionality is useful for metal free conjugations of these porphyrins to substrates, additionally avoiding the complexation of the metal by the porphyrin which is a common problem in reactions of porphyrins involving transition metal catalysts.

For affording the release of the porphyrin it is necessary to introduce labile linker bonds. It is important that these bonds are predominantly cleaved when the active agent has reached its target. Above, the synthesis of thiol-disulfide linker-containing porphyrins 5b and 6a has been described (Scheme 4). This linker moiety can be used for drug delivery and is relying on the difference of the redox potential between the cytosol and the blood stream. In the blood stream the global potential is mildly oxidative.47,51 The intracellular environment is reductive on the other hand because of the fact that the concentration of glutathione (GSH) is 103 fold higher compared to its counterpart, GSSG.52,53 In literature it is described that disulfide bonds are reduced in the cytosol, making the release of drugs possible.47,51,54,55

Another way is the pH-triggered cleavage via acetal linkers. By the time a conjugate or compound is taken up by the cell the pH drops from 7.4 to 5–6 in endosomes and even down to 4.5 in lysosomes.47,56 Yet, the acetal-linkage is stable in the blood at pH 7.4.47 Once taken up by the cell via endocytosis the linker can then be cleaved in the endosomes or lysosomes.

To evaluate the possibility to introduce an acetal-linker into the porphyrin periphery the glycerol-substituted A3B porphyrin 7[thin space (1/6-em)]57,58 was reacted with the corresponding aldehyde or dimethoxy-acetal to obtain the acetal linker-containing porphyrins 8a–c and 9 with yields between 27 and 75% (Scheme 5). Employing this method functional linker groups like epoxy, allyl, and phenolic hydroxyl were introduced. These groups make a further functionalization or linkage to a substrate possible.


image file: c6ob01551d-s5.tif
Scheme 5 Acetal formation with the A3B porphyrin 7. Reagents and conditions: (i) 4-hydroxybenzaldehyde or 4-(oxiran-2-ylmethoxy)benzaldehyde, trimethyl orthoformate, indium(III) trifluoromethane sulfonate, neat, 3–27 h, RT. (ii) 1-(Allyloxy)-4-(dimethoxymethyl)benzene, trimethyl orthoformate, indium(III) trifluoromethane sulfonate, nitromethane/THF (5/1), 72 h, RT (see Experimental section for details).

The aim was to develop a toolset for linking porphyrins to various molecular substrates. A versatile, fast and easy reaction for connecting different molecules is the CuAAC. It is commonly applied in organic,59 polymer,60 materials,61 and medicinal chemistry.62,63 Therefore, in the next step the suitability of the alkynyl-substituted porphyrins 2h,i in the CuAAC-coupling reaction was assessed (Scheme 6).


image file: c6ob01551d-s6.tif
Scheme 6 Modification of the A3B porphyrins 2h,ivia CuAAC. Reagents and conditions for all reactions: CuSO4·5H2O, L-ascorbic acid sodium salt, DMSO, 0.5–75 h, RT – 60 °C (see Experimental section for details).

The reaction of 2h with 3-azidopropanol conveys a change in the functionality from an alkyne to a hydroxyl group (in 10a) with a yield of 89%. Also the increased hydrophilicity may be favorable for a possible biological application. Glycosylated porphyrins are of great interest for the use in PDT and other fields, as they make it more specific and effective.64,65 Therefore in a test reaction alpha-D-glucose was connected to porphyrin 2i. Cancer cells show an increased uptake of glucose, which provides metabolic energy and maintains their proliferation.66,67 In various cancer cells glucose transporter proteins are over-expressed.67,68 We used 2-azido-beta-D-glucose tetraacetate which was formed in situ from acetobromo-alpha-D-glucose tetraacetate and sodium azide and reacted it with 2i to obtain the glucosylated porphyrin 11 with a yield of 17%. A large number of such CuAAC-mediated glycosylations are already described in the literature.64,65,69,70

To obtain the symmetric dimeric porphyrin 10c and the azido-porphyrin 10b (with a functionality swap from alkynyl to azido) 1,3-diazidopropane was reacted with the alkynyl-substituted porphyrin 2h. It is noteworthy that even with a high excess of 1,3-diazidopropane partial dimer formation is observed. This indicates that the reactivity of the azido-porphyrin 10b is higher compared to the 1,3-diazidopropane itself. Mannose units are known to interact with mannose receptors on the bacterial membranes which makes porphyrin–mannose conjugates possible candidates for antibacterial PDT.71–73 Therefore the azido-porphyrin 10b with the inversed end group was then further functionalized with propargyl-α-D-mannopyranoside to directly obtain the corresponding deprotected glycosylated porphyrin 10d.

Finally, the polar alkynyl-substituted porphyrin 2h was reacted with hPG19.5- or hPG116-azides 12a–d under CuAAC conditions (Scheme 7 and Table 2). By this the porphyrin-hPG19.5-conjugates 13a–c were obtained which are the first examples of conjugates combining porphyrins and the hPG carrier system. hPG is an ideal drug carrier for medical applications. The synthesis of the chemically stable hPG can easily be upscaled to the kilogram scale and the conjugate still possesses hydroxyl groups for further functionalization,33,34,37,74,75 allowing e.g. the attachment of targeting moieties.34,74 hPG is highly water soluble and tests in vitro and in vivo showed a good biocompatibility.35,36,75–77 Moreover, it shows high photostability which is advantageous with respect to its use in a photomedical application.


image file: c6ob01551d-s7.tif
Scheme 7 Functionalization of hPG with the A3B porphyrin 2hvia CuAAC. Porphyrin-, azide- and mPEG-loading of the conjugates 12a–d, 13a–c and 14a,b are given in Table 2. Reagents and conditions: (i) CuSO4·5H2O, L-ascorbic acid sodium salt, DMSO, 5 min – 75 h, RT – 40 °C; (ii) CuSO4·5H2O, L-ascorbic acid sodium salt, H2O or acetone/H2O = 11/4, v/v, 24–48 h, RT (see Experimental section for details).
Table 2 Core size and loading (porphyrin, azide and mPEG) of the hPG conjugates 12a–d, 13a–c and 14a,b
Entry Compound Core size [kDa] Porphyrin groups Azide groups mPEG groups
1 12a 19.5 ∼34
2 12b 116 ∼78
4 12c 19.5 ∼5
5 12d 19.5 ∼53
6 13a 19.5 ∼8 ∼26
7 13b 116 ∼63 ∼16
8 13c 19.5 ∼1 ∼4
9 14a 19.5 ∼1 ∼1 ∼3
10 14b 19.5 ∼8 ∼18 ∼8


hPG systems with different degrees of azide loading were used78 and reacted with different amounts of the porphyrin 2h to obtain a range of porphyrin loadings. In Table 2 the porphyrin loading is given as the approximate number of porphyrin groups. The degree of loading was determined by NMR spectroscopy by correlating the aromatic with the polyglycerol backbone protons as described in the literature.79–82

The conjugates 13a,c were further functionalized with methoxypoly(ethylene glycol) (mPEG)-propargyl ether leading to the porphyrin-mPEG-hPG19.5-conjugates 14a,b. It is known that PEGylation can be beneficial for in vivo applications as it increases the water solubility and renal clearance.83,84 Another advantage is the possible use as a carrier with so-called ‘stealth’ properties, which hides the nanoparticles from the mononuclear phagocytotic system.85 The porphyrin-hPG-conjugates 13a–c and the conjugates with additional PEGs 14a,b are examples for active substance-loaded nanocarrier systems, which may benefit from two effects: the enhanced permeability and retention (EPR)-effect86–89 and the photosensitizer-properties of the porphyrin. This makes them promising candidates for PDT.

In summary, using the nucleophilic substitution on a meso-mono-pentafluoro-substituted porphyrin carrying as additional meso-substituents three (protected) hydroxyphenyl groups a set of functionalized A3B-porphyrins suitable for the connection to carrier systems and other substrates has been prepared. The specific advantage of the present approach is that – starting from pyrrole and aldehyde – in only two steps (porphyrin condensation, nucleophilic functionalization and simultaneous deprotection) polar 3-hydroxyphenyl-substituted porphyrins with a single specific coupling site are obtained. The yields for the basic porphyrin condensation are typical for those involving the statistical condensation of two aldehydes and pyrrole (∼10%), the yields for the nucleophilic functionalization are good to very good (54–94%). As an alternative approach the selective mono-functionalization of a tetrakis(pentafluorophenyl)-substituted porphyrin has also been tested. The synthesized compounds benefit from their structural similarity with the clinically applied photosensitizer Temoporfin. For an application in the CuAAC zinc insertion in the alkynyl-substituted porphyrin was necessary as a third step. These polar porphyrins were coupled to hPG, as a prominent example of a biocompatible drug carrier system. With set of compounds at hand, we set out to investigate the photocytotoxicity of selected functionalized porphyrins and of the hPG-photosensitzer conjugates in two cancer cell lines to prove the feasibility of this approach in PDT.

Photocytotoxicity in cellular assays

The photocytotoxicity of the free porphyrin dyes 2h, 5c, 10a, 10b, and 10d was evaluated in cellular assays with human epidermoid carcinoma A-253 and squamous carcinoma CAL-27 cells (Fig. 1 and 2) (see Experimental section for details). The assays were carried out after incubation for 24 h with the photosensitizer in medium containing 10% fetal calf serum (FCS). After the 24 h incubation the medium was exchanged to ensure that only photosensitizer that has been taken up by the cells contributes to the observed effect. Both, the dark and the phototoxicity were determined at two different sensitizer concentrations (2 and 10 μmol). A white light source at a dose rate of app. 50 J cm−2 was used for irradiation. Additionally, zinc porphyrin 15, [5,10,15,20-tetrakis(3-hydroxyphenyl)porphyrinato]-zinc(II),90 was tested for comparison. Porphyrins 2h, 5c, 10a, and 10b show phototoxicity at 10 μM concentrations and in both cell lines, and exhibited a somewhat higher activity than the control sensitizer 15. At the concentration of 2 μM the porphyrins 2h, 10a, and 10b show increased phototoxicity against the cell line CAL-27. For A-253 cells the highest phototoxicity at the concentration of 2 μM is observed with porphyrin 10a. Porphyrins with terminal hydroxyl groups are described in literature to exhibit a higher phototoxicity.5 In this case for porphyrin 10a a much better efficacy compared to the control porphyrin 15 was observed. The zinc-porphyrin 10d with the mannose functionality displayed a lower toxicity and was only active at a concentration of 10 μmol. Hence, in this case neither the mannose substitution nor the concomitant increase in polarity via the additional OH groups did increase the phototoxicity of the sensitizer. None of the tested sensitizers showed dark toxicity in the CAL-27 cell line. Compounds 2h, 10a, and the control zinc porphyrin 15 showed only minor dark toxicity at the highest concentration of 10 μmol in the A253 cell line.
image file: c6ob01551d-f1.tif
Fig. 1 Photocytotoxicity of the porphyrins 2h, 5c, and 10a in cellular assays with human epidermoid carcinoma A-253 and squamous carcinoma CAL-27 cells, irradiated with a white light source (see Experimental section for details). DT: dark toxicity.

image file: c6ob01551d-f2.tif
Fig. 2 Photocytotoxicity of the porphyrins 10b, 10d, and control 15, [5,10,15,20-tetrakis(3-hydroxyphenyl)porphyrinato]-zinc(II), in cellular assays with human epidermoid carcinoma A-253 and squamous carcinoma CAL-27 cells, irradiated with a white light source (see Experimental section for details). DT: dark toxicity.

Furthermore, the photocytotoxicity of the porphyrin-hPG-conjugates without and with PEG 13a,b and 14a,b, respectively, were evaluated in the A-253 and the CAL-27 cell line (Fig. 3 and 4). As a control hPG19.5-azide 12d with approx. 53 azido groups was tested to evaluate the toxicity of the carrier polymer.


image file: c6ob01551d-f3.tif
Fig. 3 Photocytotoxicity of the porphyrin-hPG-conjugates 13a, 13b, and 14a in cellular assays with human epidermoid carcinoma A-253 and squamous carcinoma CAL-27 cells, irradiated with a white light source (see Experimental section for details). DT: dark toxicity.

image file: c6ob01551d-f4.tif
Fig. 4 Photocytotoxicity of the porphyrin-hPG conjugate 14b and the control 12d in cellular assays with human epidermoid carcinoma A-253 and squamous carcinoma CAL-27 cells, irradiated with a white light source (see Experimental section for details). DT: dark toxicity.

All of the conjugates except of the hPG19.5-azide control 12d showed phototoxicity at 10 μM concentrations in both cell lines. The highest phototoxicity was observed for the conjugate with approx. 8 porphyrin and 8 PEG groups 14b which exhibited a higher activity than the unfunctionalized zinc porphyrin 15. Presumably, the higher PEGylation of the carrier increases the solubility of the conjugate leading to a better availability of the photosensitizer.84 At the concentration of 2 μM the conjugates 13a and 14a,b show increased phototoxicity against the cell line CAL-27. For all conjugates in the two cell lines no or only minor dark toxicity was observed. The hPG19.5-azide 12d as a control does not show any significant toxicity with or without irradiation. The results show that the linkages do not impair the phototoxicity in the investigated cell lines compared to the basic porphyrin.

Conclusions

The reaction of mono-meso-pentafluorophenyl-substituted A3B-type porphyrins with various amines has been employed in the context of functionalizing porphyrins for the conjugation to carrier systems for PDT. The nucleophilic substitution with amines afforded a set of different A3B porphyrins with functional linkers i.e. alkenyl, alkynyl, amino, azido, epoxide, hydroxyl, and maleimido groups. Amide coupling of the porphyrins containing an amine functionality has been exemplified with propynoic acid and 3-(maleimido)propionic acid N-hydroxysuccinimide ester. The maleimido groups allow the linkage to certain other substrates (e.g. thiols) without the use of any catalyst. The versatility of the alkynyl-substituted A3B porphyrins for the CuAAC (Click reaction) has been demonstrated by the linkage to another porphyrin (dimer formation) and to sugar moieties. Finally for the first time porphyrins were conjugated to hPG as a biocompatible carrier system. Additionally, the synthesis of porphyrins with a cleavable linker and functional groups for further connections was established. Thus, a porphyrin with a reductively cleavable disulfide-bridge was obtained as well as porphyrins with a pH sensitive acetal linker. Overall, a toolkit for the functionalization of porphyrins with linkers for (bio-)conjugation is introduced. It could be shown that these linkages did not impair the phototoxicity in the investigated cell lines compared to the basic porphyrin which is an important prerequisite for their application in (bio-)conjugation. Cellular assays of selected zinc-porphyrins and porphyrin-hPG-conjugates showed promising phototoxicity, making their inclusion in PDT-active bioconjugates feasible.

Experimental section

Reagents

2,3,4,5,6-Pentafluorobenzaldehyde was purchased from Fluorochem. Acetobromo-alpha-D-glucose stabilized with 1% CaCO3 (98%); 3-acetoxybenzaldehyde (97%); indium(III) trifluoromethane sulfonate (99%); and pyrrole (98%) were purchased from ABCR. L-Ascorbic acid sodium salt (99%); 1,5-diaminopentane (98%); 2,3-dichloro-5,6-dicyano-1,4-benzoquinone (DDQ) (98%); dimethyl sulfoxide (DMSO) (≥99.7%) extra dry over molecular sieves; dimethylformamide (DMF) (99.8%) extra dry over molecular sieves; nitromethane (≥99%); tetrahydrofuran (THF), (99.5%), extra dry over molecular sieve, stabilized, AcroSeal®; trifluoroacetic acid (TFA) (99%); and trimethyl orthoformate (99%) were purchased from Acros Organics. N-Boc-cadaverine (≥97%); 1,4-diaminobutane (99%); dicyclohexylcarbodiimide (DCC) (99%); N,N-diisopropylethylamine (DIPEA) (Atofina EDIPA) (99%); 1-hydroxybenzotriazole hydrate (HOBt hydrate); methanol (≥99.8%); propargylamine (98%); propynoic acid (95%); and triethyl amine (≥99%) were purchased from Sigma Aldrich. Dichloromethane (DCM) (≥99%) was purchased from Fisher Chemical. Sodium acetate × 3·H2O for analysis (99.5%); sodium dihydrogen phosphate (99%) pure; and zinc acetate × 2·H2O for analysis (99.5%) were purchased from Grüssing. DMSO ROTIDRY® (≤200 ppm H2O) (≥99.5%); potassium hydroxide (≥85%) Ph. Eur. pellets; sodium chloride (≥99.5%) p. a, ACS, ISO; sodium hydroxide (≥99%); and sodium sulfate (≥99%) were purchased from Roth. Tetrahydrofuran (THF) (≥99.7%) for HPLC was purchased from VWR. 1,6-Diaminohexane (≥98%); cystamine hydrochloride (≥97%); and 3-maleimidopropionic acid N-hydroxysuccinimide ester (99%) were purchased from Alfa Aesar. 4-Hydroxybenzaldehyde (≥98%) for synthesis and sodium hydrogen phosphate (≥99.5%) for analysis were purchased from Merck. All these chemicals were used without further purification. Acetone-D6 (99.8%); CDCl3 (99.8%) stab. with silver; D2O (99.95%); CD3OD (99.8%); and THF-D8 (99.5%) were purchased from Deutero GmbH. 1,3-Diazidopropane,91 hPG19.5-azide 12a,c,d (synthesized from an hPG with Mw = 19.5 kDa and Mn = 8.4 kDa),78 hPG116-azide 12b (synthesized from an hPG with Mw = 116 kDa and Mn = 115 kDa),78,92,93 mPEG propargyl ether (average MW = 350),94 4-(oxiran-2-ylmethoxy)benzaldehyde,95 5,10,15-tris(3-acetoxyphenyl)-20-pentafluorophenylporphyrin (1a),27,48 5,10,15-tris(3-benzyloxyphenyl)-20-pentafluorophenylporphyrin (1b),27 5,10,15-tris(3-hydroxyphenyl)-20-pentafluorophenylporphyrin (1c),27,48 {5,10,15,20-tetrakis(pentafluorophenyl)porphyrinato}-zinc(II) (3),96,97 [5,10,15,20-tetrakis(3-hydroxyphenyl)porphyrinato]-zinc(II) (15),90 and 5,10,15-tris(3-hydroxyphenyl)-20-[4-(2,3-dihydroxypropoxy)tetrafluorophenyl]porphyrin (7)57,58 were prepared according to the literature or with slight modifications.

Thin-layer chromatography (TLC)

TLC analysis was performed on Merck silica gel 60 F254 precoated aluminium sheets with fluorescence indicator F254. In addition, detection of the intrinsic tetrapyrrole fluorescence was performed with UV light at 366 nm.

Column chromatography

The preparative purification of mixtures by column chromatography was conducted on silica gel, pore size 60 Å, 40–63 μm particle size, high purity containing 0.1% Ca from Fluka or MN Silica Gel 60 M, 0.04–0.063 mm/230–400 mesh, American Society for Testing (ASTM) for column chromatography from Machery-Nagel. The different eluents and the brands of the silica gel used in the synthesis are given in the individual procedures.

Dialysis

Dialysis (dialysis tubing benzoylated, avg. flat width 32 mm (1.27 in), Sigma Aldrich) was performed in 1 or 2 L beakers and the solvents were changed 3 times over a period of 24 h. The solvents used are given in the individual procedures.

NMR spectroscopy

1H, 13C, and 19F spectra were recorded on Bruker BioSpin™ AC250 (1H NMR: 250 MHz), JEOL™ ECX 400 (1H NMR: 400 MHz, 19F NMR: 376 MHz), JEOL™ ECP 500 (1H NMR: 500 MHz, 13C NMR: 126 MHz, 19F NMR: 471 MHz), and Bruker BioSpin AVANCE700 (1H NMR: 700 MHz, 13C NMR: 176 MHz) instruments. CDCl3, acetone-D6, D2O, CD3OD, and THF-D8 were used as deuterated solvents. Chemical shifts δ are given in ppm relative to tetramethylsilane (TMS) as an internal standard or relative to the resonance of the solvent (1H NMR: CDCl3: δ = 7.26 ppm, acetone-D6: δ = 2.05 ppm, D2O: δ = 4.79 ppm, CD3OD: δ = 3.31 ppm + 4.78 ppm, and THF-D8δ = 3.58 ppm + 1.73 ppm, 13C NMR: CDCl3: δ = 77.16 ppm, acetone-D6: δ = 29.84 ppm + 206.26 ppm, CD3OD: δ = 49.00 ppm, and THF-D8δ = 67.57 ppm + 25.37 ppm). All spectra were recorded at RT. Abbreviations for the signals: s (singlet), bs (broad singlet), d (doublet), t (triplet), q (quartet), quin (quintet), h (heptet), m (multiplet), dd (doublet of doublets), dt (doublet of triplets), and td (triplet of doublets).

MS spectrometry

Electrospray ionization (ESI) mass spectra were measured on an Agilent 6210 ESI-TOF from Agilent Technologies.

UV/Vis spectroscopy

The UV/Vis measurements were performed on a Specord S300 spectrometer from Analytik Jena at RT. The solvents are given in the individual procedures.

In vitro biological studies

Human epidermoid carcinoma A-253 and squamous carcinoma CAL-27 cells were grown in Dulbecco's modified eagle medium (DMEM) from cc-pro GmbH with 10% heat inactivated FCS from cc-pro GmbH, 1% penicillin (10[thin space (1/6-em)]000 IU) and streptomycin (10[thin space (1/6-em)]000 μg mL−1) from cc-pro GmbH. A stock solution (2 mM) of the PS was prepared at 4 °C in DMSO and kept in the dark. DMEM (without phenol red) with 10% FCS was used for further dilution to reach concentration 2 or 10 μM of the PS, respectively. In micro plates 2 × 104 cells per well were seeded with fresh medium (DMEM without phenol red) containing 10% FCS with 2 μM or 10 μM of the PS and incubated for 24 h. After exchange of medium (to remove any PS not taken up by the cells), the photosensitization was performed at RT with a white light source (Schott KL 200 LCD) at a dose rate of app. 50 J cm−2. The cell viability of the samples was measured with a Tecan Infinite 200 microplate reader from Tecan Group AG, Switzerland, at a wavelength of 490 nm, assessed using the XTT assays98 and the absorbance. A wavelength of 630 to 690 nm was used to measure the reference absorbance (for measuring the non-specific readings).

Recrystallization

Recrystallization of the porphyrinoids was performed by dissolving the product in the minimum amount of solvent (e.g. DCM) and layering it with a 3-fold excess of the anti-solvent (e.g. methanol/water = 9/1, v/v).

Melting point (m.p.) measurements

The m.p. measurements were performed on a Thermovar m.p. microscope from Reichert.

General synthesis of the zinc-porphyrins 1d, 2h, 2i, and 5c

In a flask with magnetic stirrer the porphyrin 1c, 2f, 2g, or 5b was dissolved in methanol or a DCM/methanol mixture. A point of a spatula of sodium acetate and zinc acetate dihydrate was added to the stirred solution. The solution was stirred for 0.5 to 18 h at RT. The crude product was diluted with ethyl acetate or DCM and washed with H2O. Afterwards the organic layer was dried over Na2SO4 and the solution was evaporated to dryness. The crude product was purified by column chromatography and/or recrystallization from DCM/n-hexane to obtain the corresponding zinc-porphyrins 1d, 2h, 2i, and 5c. Detailed experimental conditions are given in the ESI. The products were analyzed by NMR, MS, and UV/Vis spectroscopy.

General synthesis of the porphyrins 2a, 2b, 2c, 2d, 2e, 2f, and 4 using the nucleophilic aromatic substitution with amines

In a flask with magnetic stirrer porphyrin 1a or 1c was dissolved in anhydrous DMSO or DMSO/THF mixture under argon. To the stirred solution the amine was added. The solution was stirred at 83 to 100 °C for 0.5 to 4 h. The crude product was diluted with ethyl acetate or DCM and washed with H2O and/or saturated NaCl-solution. Afterwards the organic layer was dried over Na2SO4. The crude product was evaporated to dryness and the remaining residue was purified by column chromatography and recrystallization to obtain the porphyrin products 2a, 2b, 2c, 2d, 2e, 2f and 4. Detailed experimental conditions are given in the ESI. The products were analyzed by NMR, MS, and UV/Vis spectroscopy.
5,10,15-Tris(3-benzyloxyphenyl)-20-[4-(prop-2-ynylamino)tetrafluorophenyl]porphyrin (2g). In a 10 mL flask with magnetic stirrer 5,10,15-tris(3-benzyloxyphenyl)-20-pentafluorophenylporphyrin (1b) (156 mg, 152 μmol) was dissolved in 3 mL of anhydrous THF (Acros) under argon. 3 mL of anhydrous DMSO (Roth) were added. The THF was evaporated in vacuo as long as the porphyrin stayed in solution. Propargylamine (98%, 160 μL, 2.44 mmol) was added and the solution was stirred at 100 °C for 7 h. The crude product was diluted with 100 mL of DCM and washed twice with 100 mL of H2O. Afterwards the organic layer was dried over Na2SO4. The crude product was evaporated to dryness and the remaining residue was purified by column chromatography (DCM/n-hexane = 3/1, v/v, Machery-Nagel) and recrystallization from DCM/methanol to obtain 5,10,15-tris(3-benzyloxyphenyl)-20-[4-(prop-2-ynylamino)tetrafluorophenyl]porphyrin (2g) (125 mg, 118 μmol, 78% yield) as a purple solid.

1H NMR (CDCl3, 700 MHz): δ = 8.94 (d, 3J(H,H) = 4.2 Hz, 2H, 2,18-β), 8.89–8.83 (m, 6H, 3,7,8,12,13,17-β), 7.88 (s, 3H, Ar), 7.86–7.82 (m, 3H, Ar), 7.67 (t, 3J(H,H) = 7.8 Hz, 3H, Ar), 7.53 (d, 3J(H,H) = 7.6 Hz, 6H, Ar), 7.44–7.39 (m, 9H, Ar), 7.38–7.32 (m, 3H, Ar), 5.27 (s, 6H, OCH2), 4.49–4.43 (m, 3H, NHCH2 + ArF-NH), 2.50 (s, 1H, C[triple bond, length as m-dash]CH) −2.78 ppm (s, 2H, pyrrole-NH). 13C NMR (CDCl3, 126 MHz): δ = 157.27, 147.56, 146.17, 143.46, 143.30, 138.70, 137.34, 137.04, 131.66, 128.80, 128.19, 128.15, 127.80, 121.66, 121.34, 120.39, 114.91, 110.70, 102.40, 80.43, 73.03, 70.43, 36.13. 19F NMR (CDCl3, 471 MHz): δ = −139.78 (dd, 3J(F,F) = 22.0 Hz; 4J(F,F) = 8.3 Hz, 2F, m-ArF), −158.89–(−159.30) ppm (m, 2F, o-ArF). m.p.: 80 °C. HRMS (ESI): calc. for C68H48F4N5O3+ ([M + H]+): 1058.3693 found: 1058.3651. UV/Vis (DCM): λmax (ε [M−1 cm−1]) = 645 (3000), 589 (6000), 548 (6000), 514 (18[thin space (1/6-em)]000), 419 nm (338[thin space (1/6-em)]000).

5,10,15-Tris(3-hydroxyphenyl)-20-[4-(N-4-propyneamidobutylamino)tetrafluorophenyl]porphyrin (5a). In a 10 mL flask with magnetic stirrer propynoic acid (95%, 3.00 μL, 3.40 mg, 46.1 μmol), HOBt hydrate (7.40 mg, 54.8 μmol), DCC (99%, 17.3 mg, 83.0 μmol) was dissolved in 1 mL THF (VWR). The solution was stirred for 10 min at RT. To the stirred solution 5,10,15-tris(3-hydroxyphenyl)-20-[4-(4-aminobutylamino)tetrafluorophenyl]porphyrin (2b) (40.1 mg, 48.9 μmol) was added. The solution was stirred at RT for 2 h. The crude product was diluted with 150 mL of ethyl acetate and washed three times with 50 mL of H2O. Afterwards the organic layer was dried over Na2SO4. The crude product was evaporated to dryness and the remaining residue was purified by column chromatography (DCM/methanol = 94/6, v/v, Fluka) to obtain 5,10,15-tris(3-hydroxyphenyl)-20-[4-(N-4-propyneamidobutylamino)tetrafluorophenyl]porphyrin (5a) (13.9 mg, 15.9 μmol, 33% yield) as a purple solid. The relatively low yield is due to the fact that the product partly decomposed during workup. Also the final product exhibited a low stability in solution.

1H NMR (THF-D8, 500 MHz): δ = 8.99–8.84 (m, 11H, β + 5,10,15-meso-3-Ar-OH), 7.90 (s, 1H, NHC(O)), 7.69–7.61 (m, 6H, 5,10,15-meso-2,6-Ar), 7.58–7.51 (m, 3H, 5,10,15-meso-5-Ar), 7.22–7.18 (m, 3H, 5,10,15-meso-4-Ar), 5.81 (s, 1H, ArF-NH), 3.69 (q, 3J(H,H) = 6.6 Hz, 2H, ArF-NHCH2), 3.37 (s, 1H, C[triple bond, length as m-dash]CH), 3.35 (q, 3J(H,H) = 6.7 Hz, 2H, CH2NHC(O)), 1.91–1.84 (m, 2H, ArF-NHCH2CH2), 1.80–1.74 (m, 2H, CH2CH2NHC(O)), −2.73 ppm (s, 2H, pyrrole-NH). 13C NMR (THF-D8, 126 MHz): δ = 157.32, 157.28, 152.59, 149.08, 147.15, 144.29, 144.16, 138.93, 137.08, 130.49, 128.35, 128.30, 127.06, 123.05, 122.28, 121.37, 115.81, 103.59, 79.52, 73.01, 67.99, 54.96, 46.25, 39.92, 30.71, 29.36, 27.79, 25.86 ppm. 19F NMR (THF-D8, 376 MHz): δ = −142.72–(−143.27) (m, 2F, m-ArF), −162.78–(−163.07) ppm (m, 2F, o-ArF). m.p.: >230 °C. HRMS (ESI): calc. for C51H37F4N6O4+ ([M + H]+): 873.2807; found: 873.2806. UV/Vis (acetone): λmax (ε [M−1 cm−1]) = 645 (3000), 592 (5000), 546 (6000), 512 (16[thin space (1/6-em)]000), 416 nm (203[thin space (1/6-em)]000).

5,10,15-Tris(3-hydroxyphenyl)-20-[2,3,5,6-tetrafluoro-4-(N-(2-((2-aminoethyl)disulfanyl)ethylpropyneamido))-phenyl]porphyrin (5b). In a 10 mL flask with magnetic stirrer DCC (99%, 16.0 mg, 76.7 μmol), propynoic acid (95%, 4.82 μL, 73.9 μmol), and HOBt hydrate (12.0 mg, 88.8 μmol) were dissolved in 1 mL of THF (VWR) and stirred for 10 min at RT. 5,10,15-Tris(3-hydroxyphenyl)-20-[2,3,5,6-tetrafluoro-4-(N-(2-((2-aminoethyl)disulfanyl)ethylamino))phenyl]porphyrin (2a) (69.0 mg, 78.0 μmol) was added and the solution was stirred for 2 h at RT. The crude product was dissolved in 100 mL of ethyl acetate and washed three times with 50 mL of H2O. Afterwards the organic layer was dried over Na2SO4 and the solution was evaporated to dryness. The crude product was purified by column chromatography (DCM/methanol = 85/15, v/v, Machery-Nagel) and recrystallization from DCM/n-hexane to obtain 5,10,15-tris(3-hydroxyphenyl)-20-[2,3,5,6-tetrafluoro-4-(N-(2-((2-aminoethyl)disulfanyl)ethylpropyneamido))phenyl]porphyrin (5b) (56.0 mg, 59.8 μmol, 77% yield) as a purple solid.

1H NMR (acetone-D6, 700 MHz): δ = 9.13–9.10 (bs, 2H, 2,18-β), 9.04–9.01 (bs, 2H, 3,17-β), 9.00–8.95 (m, 7H, 7,8,12,13-β + 5,10,15-meso-3-Ar-OH), 8.10–8.07 (bs, 1H, NHC(O)), 7.76 (d, 4J(H,H) = 2.1 Hz, 2H, 5,15-meso-2-Ar), 7.75 (d, 4J(H,H) = 2.1 Hz, 1H, 10-meso-2-Ar), 7.73 (d, 3J(H,H) = 7.7 Hz, 2H, 5,15-meso-6-Ar), 7.72 (d, 3J(H,H) = 8.7 Hz, 1H, 10-meso-6-Ar), 7.66–7.61 (m, 3H, 5,10,15-meso-5-Ar), 7.33 (dd, 3J(H,H) = 8.5, 4J(H,H) = 2.3 Hz, 3H, 5,10,15-meso-4-Ar), 5.91 (t, 3J(H,H) = 7.1 Hz, 1H, ArF-NH), 4.04 (q, 3J(H,H) = 6.9 Hz, 2H, ArF-NHCH2), 3.68 (q, 3J(H,H) = 6.6 Hz, 2H, CH2NHC(O)), 3.53 (s, 1H, C[triple bond, length as m-dash]CH), 3.26 (t, 3J(H,H) = 6.7 Hz, 2H, ArF-NHCH2CH2), 3.02 (t, 3J(H,H) = 6.8 Hz, 2H, CH2CH2NHC(O)), −2.75 ppm (s, 2H, pyrrole-NH). 13C NMR (acetone-D6, 176 MHz): δ = 156.85, 156.80, 152.91, 148.56, 147.22, 143.94, 143.80, 138.96, 137.50, 132.13, 129.94, 128.66, 128.61, 127.19, 127.14, 122.84, 122.80, 122.34, 121.38, 115.98, 108.24, 103.56, 78.69, 74.46, 45.44, 39.57, 39.43, 37.95 ppm. 19F NMR (acetone-D6, 376 MHz): δ = −143.11 (d, 3J(F,F) = 21.0 Hz, 2F, m-ArF), −161.79 ppm (d, 3J(F,F) = 18.9 Hz, 2F, o-ArF). m.p.: >230 °C. HRMS (ESI): calc. for C51H37F4N6O4S2+ ([M + H]+): 937.2254 found: 937.2294. UV/Vis (ethanol): λmax (ε [M−1 cm−1]) = 645 (3000), 589 (6000), 547 (7000), 512 (18[thin space (1/6-em)]000), 416 nm (329[thin space (1/6-em)]000).

5,10,15-Tris(3-hydroxyphenyl)-20-[4-((2-((2-((3-maleimidyl)propanamido)ethyl)disulfanyl)ethyl)amino)tetrafluorophenyl]porphyrin (6a). In a 10 mL flask with magnetic stirrer under argon 5,10,15-tris(3-hydroxyphenyl)-20-[4-((2-((2-aminoethyl)disulfanyl)ethyl)amino)tetrafluorophenyl]porphyrin (2a) (122 mg, 138 μmol) was dissolved in 1.5 mL of anhydrous DMF. 3-(Maleimido)propionic acid N-hydroxysuccinimide ester (99%, 47.1 mg, 177 μmol) was added and the solution was stirred for 1 h at RT. The reaction mixture was diluted with 100 mL ethyl acetate and washed four times with 150 mL H2O. The organic layer was dried over Na2SO4 and the solvent was evaporated in vacuo. The crude product was purified by column chromatography (DCM/methanol = 95/5, v/v, Fluka). The product was recrystallized from n-hexane to obtain 5,10,15-tris(3-hydroxyphenyl)-20-[4-((2-((2-((3-maleimidyl)propanamido)ethyl)disulfanyl)ethyl)amino)tetrafluorophenyl]porphyrin (6a) (116 mg, 112 μmol, 81% yield).

1H NMR (THF-D8, 500 MHz): δ = 9.02–8.85 (bm, 8H, β), 8.75–8.66 (m, 3H, 5,10,15-meso-3-Ar-OH), 7.72–7.61 (m, 6H, 5,10,15-meso-2,6-Ar), 7.55 (t, 3J(H,H) = 7.8 Hz, 3H, 5,10,15-meso-5-Ar), 7.53–7.44 (m, 1H, NHC(O)), 7.26–7.14 (m, 3H, 5,10,15-meso-4-Ar), 6.74 (s, 2H, HC[double bond, length as m-dash]CH), 6.11–6.03 (bs, 1H, ArF-NH), 3.98 (d, 3J(H,H) = 7.3 Hz, 2H, ArF–NHCH2), 3.83–3.69 (m, 2H, CH2N), 3.53 (t, 3J(H,H) = 6.2 Hz, 2H, CH2NHC(O)), 3.19 (t, 3J(H,H) = 6.8 Hz, 2H, ArF-NHCH2CH2S), 2.90 (t, 3J(H,H) = 6.7 Hz, 2H, SCH2CH2NHC(O)), 2.50–2.36 (m, 2H, C(O)CH2), −2.72 ppm (s, 2H, pyrrole-NH). 13C NMR (THF-D8, 126 MHz): δ = 171.04, 170.21, 157.08, 157.04, 148.77, 146.84, 144.03, 143.89, 138.88, 136.97, 134.86, 131.60, 129.65, 128.11, 128.05, 126.80, 122.81, 122.08, 121.16, 115.58, 108.21, 103.20, 45.44, 39.68, 39.07, 38.45, 34.93, 34.91 ppm. 19F NMR (THF-D8, 471 MHz): δ = −142.56–(−142.86) (m, 2F, m-ArF), −162.24–(−162.47) ppm (m, 2F, o-ArF). m.p.: 185 °C. HRMS (ESI): calc. for C55H42F4N7O6S2+ ([M + H]+): 1036.2569 found: 1036.2588. UV/Vis (methanol): λmax (ε [M−1 cm−1]) = 645 (3000), 588 (5000), 546 (6000), 512 (16[thin space (1/6-em)]000), 415 nm (229[thin space (1/6-em)]000).

5,10,15-Tris(3-hydroxyphenyl)-20-[4-((((5-maleimidyl)propanamido)pentyl)amino)tetrafluorophenyl]porphyrin (6b). In a 10 mL flask with magnetic stirrer under argon 5,10,15-tris(3-hydroxyphenyl)-20-[4-(5-aminopentylamino)tetrafluorophenyl]porphyrin (2c) (46.1 mg, 55.2 μmol) was dissolved in 1.5 mL of anhydrous DMF. 3-(Maleimido)propionic acid N-hydroxysuccinimide ester (99%, 19.8 mg, 73.6 μmol) was added and the solution was stirred for 1 h at RT. The reaction mixture was diluted with 100 mL ethyl acetate and washed four times with 150 mL H2O. The organic layer was dried over Na2SO4 and the solvent was evaporated in vacuo. The crude product was purified by column chromatography (DCM/methanol = 95/5, v/v, Fluka). The product was recrystallized from n-hexane to obtain 5,10,15-tris(3-hydroxyphenyl)-20-[4-((((5-maleimidyl)propanamido)pentyl)amino)tetrafluorophenyl]porphyrin (6b) (35.3 mg, 35.8 μmol, 65% yield).

1H NMR (THF-D8, 500 MHz): δ = 9.02–8.85 (m, 8H, β), 8.76 (s, 3H, 5,10,15-meso-3-Ar-OH), 7.71–7.62 (m, 6H, 5,10,15-meso-2,6-Ar), 7.55 (t, 3J(H,H) = 7.8 Hz, 3H, 5,10,15-meso-5-Ar), 7.24–7.19 (m, 3H, 5,10,15-meso-4-Ar), 7.17 (t, 3J(H,H) = 5.0 Hz, 1H, NHC(O)), 6.76 (s, 2H, HC[double bond, length as m-dash]CH), 5.75 (s, 1H, ArF-NH), 3.79–3.72 (m, 2H, C(O)CH2CH2), 3.66 (q, 3J(H,H) = 6.6 Hz, 2H, ArF-NHCH2), 3.24 (q, 3J(H,H) = 6.5 Hz, 2H, CH2NHC(O)), 2.45–2.38 (m, 2H, C(O)CH2), 1.87 (quin, 3J(H,H) = 7.4 Hz, 2H, ArF-NHCH2CH2), 1.65–1.51 (m, 4H, ArF-NHCH2CH2CH2CH2), −2.72 ppm (s, 2H, pyrrole-NH). 13C NMR (THF-D8, 126 MHz): δ = 171.28, 169.83, 157.27, 157.23, 149.02, 147.10, 144.32, 144.19, 135.10, 128.36, 128.31, 127.15, 127.12, 123.05, 122.25, 121.35, 115.79, 103.65, 46.56, 39.76, 35.25, 35.16, 31.76, 30.66, 25.86 ppm. 19F NMR (THF-D8, 376 MHz): δ = −142.87–(−143.22) (m, 2F, m-ArF), −162.24 ppm (d, 3J(F,F) = 14.2 Hz, 2F, o-ArF). m.p.: >300 °C. HRMS (ESI): calc. for C56H44F4N7O6+ ([M + H]+): 986.3284 found: 986.3329. UV/Vis (methanol): λmax (ε [M−1 cm−1]) = 645 (3000), 588 (6000), 546 (7000), 513 (19[thin space (1/6-em)]000), 415 nm (257[thin space (1/6-em)]000).

5,10,15-Tris(3-hydroxyphenyl)-20-[4-((((6-maleimidyl)propanamido)hexyl)amino)tetrafluorophenyl]porphyrin (6c). In a 10 mL flask with magnetic stirrer under argon 5,10,15-tris(3-hydroxyphenyl)-20-[4-(6-aminohexylamino)tetrafluorophenyl]porphyrin (2d) (78.6 mg, 92.6 μmol) was dissolved in 1.5 mL of anhydrous DMF. 3-(Maleimido)propionic acid N-hydroxysuccinimide ester (99%, 30.2 mg, 112 μmol) was added and the solution was stirred for 1 h at RT. The reaction mixture was diluted with 100 mL ethyl acetate and washed four times with 150 mL H2O. The organic layer was dried over Na2SO4 and the solvent was evaporated in vacuo. The crude product was purified by column chromatography (DCM/methanol = 95/5, v/v, Fluka). The product was recrystallized from n-hexane to obtain 5,10,15-tris(3-hydroxyphenyl)-20-[4-((((6-maleimidyl)propanamido)hexyl)amino)tetrafluorophenyl]porphyrin (6c) (62.8 mg, 62.8 μmol, 68% yield).

1H NMR (THF-D8, 500 MHz): δ = 9.00–8.88 (m, 8H, β), 8.75–8.88 (m, 3H, 5,10,15-meso-3-Ar-OH), 7.69–7.62 (m, 6H, 5,10,15-meso-2,6-Ar), 7.55 (t, 3J(H,H) = 7.8 Hz, 3H, 5,10,15-meso-5-Ar), 7.20 (dd, 3J(H,H) = 7.9, 4J(H,H) = 2.3 Hz, 3H, 5,10,15-meso-4-Ar), 7.13 (t, 3J(H,H) = 6.0 Hz, 1H, NHC(O)), 6.75 (s, 2H, HC[double bond, length as m-dash]CH), 5.77 (t, 3J(H,H) = 6.0 Hz, 1H, ArF-NH), 3.78–3.70 (m, 2H, C(O)CH2CH2), 3.66 (q, 3J(H,H) = 7.3 Hz, 2H, ArF-NHCH2), 3.20 (q, 3J(H,H) = 6.5 Hz, 2H, CH2NHC(O)), 2.44–2.36 (m, 2H, C(O)CH2), 1.85 (quin, 3J(H,H) = 7.4 Hz, 2H, ArF-NHCH2CH2), 1.61–1.51 (m, 4H, ArF-NHCH2CH2CH2CH2CH2), 1.51–1.41 (m, 2H, ArF-NHCH2CH2CH2CH2), −2.71 ppm (s, 2H, pyrrole-NH). 13C NMR (THF-D8, 126 MHz): δ = 171.26, 169.74, 157.25, 157.21, 149.15, 147.10, 144.33, 144.19, 135.08, 130.53, 128.37, 128.32, 127.17, 127.14, 123.05, 122.24, 121.34, 115.78, 103.65, 46.46, 39.77, 35.24, 35.14, 32.09, 30.85, 30.70, 27.68, 27.51 ppm. 19F NMR (THF-D8, 471 MHz): δ = −142.88–(−143.17) (m, 2F, m-ArF), −163.09 ppm (d, 3J(F,F) = 15.9 Hz, 2F, o-ArF). m.p.: 181 °C. HRMS (ESI): calc. for C57H46F4N7O6+ ([M + H]+): 1000.3440 found: 1000.3460. UV/Vis (methanol): λmax (ε [M−1 cm−1]) = 645 (4000), 588 (7000), 545 (8000), 512 (20[thin space (1/6-em)]000), 415 nm (263[thin space (1/6-em)]000).

(±)-5,10,15-Tris(3-hydroxyphenyl)-20-[4-((2-methoxy-1,3-dioxolan-4-yl)methoxy)tetrafluorophenyl]porphyrin (8a). In a sample tube with magnetic stirrer 5,10,15-tris(3-hydroxyphenyl)-20-[4-(2,3-dihydroxypropoxy)tetrafluorophenyl]porphyrin (7) (31.2 mg, 37.8 μmol), 4-hydroxybenzaldehyde (98%, 58.8 mg, 472 μmol), trimethyl orthoformate (99%, 79 μL, 720 μmol), and indium(III) trifluoromethane sulfonate (99%, 2.8 mg, 4.9 μmol) were mixed and stirred neat for 3 h. The reaction mixture was diluted with 100 mL ethyl acetate and washed three times with 100 mL phosphate buffer (100 mM, pH 8). The organic layer was dried over Na2SO4 and the solvent was evaporated in vacuo. The crude product was purified by column chromatography (n-hexane/acetone = 3/2, v/v, Fluka) to obtain (±)-5,10,15-tris(3-hydroxyphenyl)-20-[4-((2-methoxy-1,3-dioxolan-4-yl)methoxy)tetrafluorophenyl]porphyrin (8a) (20.1 mg, 23.2 μmol, 61% yield).

1H NMR (acetone-D6, 500 MHz): δ = 9.10 (d, 3J(H,H) = 4.0 Hz, 2H, 2,18-β), 9.03 (d, 3J(H,H) = 4.4 Hz, 2H, 3,17-β), 8.98 (d, 3J(H,H) = 2.2 Hz, 4H, 7,8,12,13-β), 9.00–8.87 (bs, 3H, 5,10,15-meso-3-Ar-OH), 7.78–7.75 (m, 3H, 5,10,15-meso-2-Ar), 7.75–7.71 (m, 3H, 5,10,15-meso-6-Ar), 7.66–7.61 (m, 3H, 5,10,15-meso-5-Ar), 7.36–7.32 (m, 3H, 5,10,15-meso-4-Ar), 5.97, 5.92 (s, 1H, acetal-H), 4.91–4.64 (m, 3H), 4.39–4.33 (m, 1H), 4.17–4.07 (m, 1H), 3.41, 3.36 (s, 3H, CH3), −2.74 ppm (s, 2H, pyrrole-NH). 13C NMR (acetone-D6, 126 MHz): δ = 156.83, 156.77, 148.68, 146.77, 143.87, 143.69, 141.25, 139.36, 132.35, 128.68, 128.62, 127.20, 127.16, 122.83, 122.65, 121.45, 117.44, 1.10, 116.00, 115.83, 102.22, 76.82, 75.84, 75.75, 75.12, 66.14, 65.97, 51.54, 51.15 ppm. 19F NMR (acetone-D6, 471 MHz): δ = −141.47–(−141.71) (m, 2F, m-ArF), −158.70–(−158.92) ppm (m, 2F, o-ArF). m.p.: >300 °C. HRMS (ESI): calc. for C49H34F4N4O7+ ([M + H]+): 867.2442 found: 867.2456. UV/Vis (ethanol): λmax (ε [M−1 cm−1]) = 644 (2000), 588 (6000), 545 (6000), 511 (19[thin space (1/6-em)]000), 415 nm (383[thin space (1/6-em)]000).

(±)-5,10,15-Tris(3-hydroxyphenyl)-20-[4-((2-(4-hydroxyphenyl)-1,3-dioxolan-4-yl)methoxy)tetrafluorophenyl]porphyrin (8b). In a sample tube with magnetic stirrer 4-hydroxybenzaldehyde (98%, 80.3 mg, 644 μmol), trimethyl orthoformate (99%, 51 μL, 460 μmol), and indium(III) trifluoromethane sulfonate (99%, 4.2 mg, 7.4 μmol) were mixed and stirred neat for 3 h. 5,10,15-Tris(3-hydroxyphenyl)-20-[4-(2,3-dihydroxypropoxy)tetrafluorophenyl]porphyrin (7) (30.0 mg, 36.4 μmol) was added and the mixture was stirred for another 2 h. The reaction was quenched with triethyl amine (99%, 500 μL, 3.55 mmol). The reaction mixture was diluted with 100 mL ethyl acetate and washed three times with 100 mL phosphate buffer (100 mM, pH 8). The organic layer was dried over Na2SO4 and the solvent was evaporated in vacuo. The crude product was purified by column chromatography (n-hexane/acetone = 3/2, v/v, Fluka) to obtain (±)-5,10,15-tris(3-hydroxyphenyl)-20-[4-((2-(4-hydroxyphenyl)-1,3-dioxolan-4-yl)methoxy)tetrafluorophenyl]porphyrin (8b) (25.3 mg, 27.2 μmol, 75% yield).

1H NMR (acetone-D6, 500 MHz): δ = 9.11–8.88 (bm, 11H, β + 5,10,15-meso-3-Ar-OH), 8.74–8.47 (bs, 1H, acetal-4-Ar-OH), 7.79–7.71 (m, 6H, 5,10,15-meso-2,6-Ar), 7.637 (t, 3J(H,H) = 7.8 Hz, 2H, 5,15-meso-5-Ar), 7.630 (t, 3J(H,H) = 7.9 Hz, 1H, 10-meso-5-Ar), 7.49, 7.43 (d, 3J(H,H) = 8.1, 8.7 Hz, 2H, acetal-2,6-Ar), 7.34 (d, 3J(H,H) = 8.4 Hz, 3H, 5,10,15-meso-4-Ar), 6.91 (d, 3J(H,H) = 8.4 Hz, 2H, acetal-3,5-Ar), 6.05, 5.85 (s, 1H, acetal-H), 4.88–4.72 (m, 3H), 4.53–4.09 (m, 2H), −2.75 ppm (s, 2H, pyrrole-NH). 13C NMR (acetone-D6, 126 MHz): δ = 159.44, 159.26, 156.86, 156.82, 148.74, 146.82, 143.93, 143.74, 141.29, 139.58, 132.79, 130.05, 129.49, 129.39, 129.14, 128.71, 128.64, 127.28, 127.20, 122.90, 122.85, 122.68, 121.60, 116.04, 115.91, 115.88, 105.72, 104.98, 102.30, 76.37, 76.08, 75.87, 75.77, 67.75, 67.51 ppm. 19F NMR (acetone-D6, 471 MHz): δ = −141.47–(−141.71) (m, 2F, m-ArF), −158.70–(−158.92) ppm (m, 2F, o-ArF). m.p.: 60 °C. HRMS (ESI): calc. for C54H37F4N4O7+ ([M + H]+): 929.2598 found: 929.2632. UV/Vis (DCM): λmax (ε [M−1 cm−1]) = 645 (2000), 589 (4000), 548 (4000), 514 (12[thin space (1/6-em)]000), 418 nm (220[thin space (1/6-em)]000).

(±)-5,10,15-Tris(3-hydroxyphenyl)-20-[4-((2-(4-(oxiran-2-ylmethoxy)phenyl)-1,3-dioxolan-4-yl)methoxy)tetrafluorophenyl]porphyrin (8c). In a sample tube with magnetic stirrer 4-(oxiran-2-ylmethoxy)benzaldehyde (92.4 mg, 519 μmol), trimethyl orthoformate (99%, 39 μL, 350 μmol), and indium(III) trifluoromethane sulfonate (99%, 4.2 mg, 7.4 μmol) were mixed and stirred neat for 3 h. 5,10,15-Tris(3-hydroxyphenyl)-20-[4-(2,3-dihydroxypropoxy)tetrafluorophenyl]porphyrin (7) (32.1 mg, 38.9 μmol) and 2 drops of DCM were added and the mixture was stirred for another 24 h. The reaction was quenched with triethyl amine (99%, 100 μL, 710 μmol). The reaction mixture was diluted with 100 mL ethyl acetate and washed three times with 100 mL phosphate buffer (100 mM, pH 8). The organic layer was dried over Na2SO4 and the solvent was evaporated in vacuo. The crude product was purified by column chromatography (n-hexane/acetone = 1/1, v/v, Fluka) followed by a second column chromatography (n-hexane/acetone = 3/2, v/v, Fluka) to obtain (±)-5,10,15-tris(3-hydroxyphenyl)-20-[4-((2-(4-(oxiran-2-ylmethoxy)phenyl)-1,3-dioxolan-4-yl)methoxy)tetrafluorophenyl]porphyrin (8c) (10.4 mg, 10.6 μmol, 27% yield).

1H NMR (acetone-D6, 500 MHz): δ = 9.10–8.89 (bm, 11H, β + 5,10,15-meso-3-Ar-OH), 7.77–7.70 (m, 6H, 5,10,15-meso-2,6-Ar), 7.67–7.61 (m, 3H, 5,10,15-meso-5-Ar), 7.57, 7.52 (d, 3J(H,H) = 8.6, 8.6 Hz, 2H, acetal-2,6-Ar), 7.36–7.31 (m, 3H, 5,10,15-meso-4-Ar), 7.04, 7.03 (d, 3J(H,H) = 8.7, 8.7 Hz, 2H, acetal-3,5-Ar), 6.07, 5.89 (s, 1H, acetal-H), 4.90–4.73 (m, 3H), 4.54–3.17 (m, 5.5H), 2.75–2.54 (m, 1.5H), −2.76–(−2.80) ppm (m, 2H, pyrrole-NH). 13C NMR (acetone-D6, 126 MHz): δ = 159.84, 159.69, 156.06, 156.00, 143.09, 142.92, 142.91, 131.71, 131.66, 130.80, 130.25, 128.55, 128.28, 127.91, 127.84, 126.44, 126.38, 122.06, 122.01, 121.86, 121.85, 120.78, 120.76, 115.23, 114.96, 114.37, 114.33, 104.66, 104.64, 103.86, 75.48, 75.30, 75.16, 75.14, 69.37, 69.35, 69.32, 69.26, 66.93, 66.71, 49.76, 49.65, 43.61, 43.51 ppm. 19F NMR (acetone-D6, 471 MHz): δ = −141.31–(−142.08) (m, 2F, m-ArF), −158.48–(−159.15) ppm (m, 2F, m-ArF). m.p.: >300 °C. HRMS (ESI): calc. for C54H41F4N4O8+ ([M + H]+): 985.2861 found: 985.2851. UV/Vis (acetone): λmax (ε [M−1 cm−1]) = 644 (2000), 588 (5000), 545 (5000), 511 (13[thin space (1/6-em)]000), 415 nm (243[thin space (1/6-em)]000).

(±)-5,10,15-Tris(3-hydroxyphenyl)-20-[4-((2-(4-(allyloxy)phenyl)-1,3-dioxolan-4-yl)methoxy)tetrafluorophenyl]porphyrin (9). In a 10 mL flask with magnetic stirrer 1-(allyloxy)-4-(dimethoxymethyl)benzene (33.0 mg, 158 μmol), 5,10,15-tris(3-hydroxyphenyl)-20-[4-(2,3-dihydroxypropoxy)tetrafluorophenyl]porphyrin (7) (80.3 mg, 97.4 μmol), and indium(III) trifluoromethane sulfonate (99%, 6.4 mg, 11 μmol) were dissolved in 5 mL of nitromethane. After 24 h 1 mL of dry THF (Acros) was added and the reaction mixture was stirred for another 24 h. 1-(Allyloxy)-4-(dimethoxymethyl)benzene (275 mg, 1.32 mmol) and indium(III) trifluoromethane sulfonate (99%, 6.6 mg, 12 μmol) were added. After 3 d the reaction was completed. The reaction mixture was diluted with 50 mL methanol/triethyl amine (99[thin space (1/6-em)]:[thin space (1/6-em)]1) and filtered over silica gel. The product was recrystallized from DCM/(methanol/H2O 4[thin space (1/6-em)]:[thin space (1/6-em)]1 + NH3 (pH 8)) to obtain (±)-5,10,15-tris(3-hydroxyphenyl)-20-[4-((2-(4-(allyloxy)phenyl)-1,3-dioxolan-4-yl)methoxy)tetrafluorophenyl]porphyrin (9) (52.5 mg, 54.2 μmol, 56% yield).

1H NMR (acetone-D6, 700 MHz): δ = 9.10–8.83 (bm, 11H, β + 5,10,15-meso-3-Ar-OH), 7.80–7.69 (m, 6H, 5,10,15-meso-2,6-Ar), 7.66–7.61 (m, 3H, 5,10,15-meso-5-Ar), 7.55, 7.51 (d, 3J(H,H) = 8.6, 8.5 Hz, 2H, acetal-2,6-Ar), 7.34 (d, 3J(H,H) = 8.4 Hz, 3H, 5,10,15-meso-4-Ar), 7.012, 7.006 (d, 3J(H,H) = 8.5, 8.6 Hz, 2H, acetal-3,5-Ar), 6.07, 5.88 (s, 1H, acetal-H), 6.11–6.05, 5.97–5.89 (m, 1H, CH[double bond, length as m-dash]CH2), 5.46–5.38, 5.29–5.21, 5.11–5.05 (m, 2 H, CH[double bond, length as m-dash]CH2), 4.90–4.72 (m, 3H), 4.61 (d, 3J(H,H) = 5.2 Hz, 1H, CH2CH[double bond, length as m-dash]), 4.55–4.41 (m, 1.5H), 4.30 (d, 3J(H,H) = 6.1 Hz, 1H, CH2CH[double bond, length as m-dash]), 4.16–4.06 (m, 0.5H), −2.75–(−2.76) ppm (m, pyrrole-NH). 13C NMR (acetone-D6, 176 MHz): δ = 160.59, 160.44, 156.85, 156.80, 148.43, 147.06, 143.92, 143.74, 142.95, 141.57, 139.62, 134.64, 134.47, 131.36, 130.79, 129.28, 129.03, 128.71, 128.64, 127.26, 127.20, 122.87, 122.83, 122.69, 122.66, 122.65, 121.60, 121.58, 121.57, 117.45, 117.37, 116.03, 115.29, 115.25, 105.51, 104.73, 102.28, 76.29, 76.12, 76.10, 76.07, 75.95, 69.32, 69.26, 67.74, 67.53, 49.78 ppm. 19F NMR (acetone-D6, 471 MHz): δ = −141.54–(−141.89) (m, 2F, m-ArF), −158.58–(−158.87) ppm (m, 2F, m-ArF). m.p.: 140–162 °C. HRMS (ESI): calc. for C57H41F4N4O7+ ([M + H]+): 969.2911 found: 969.2915. UV/Vis (acetone): λmax (ε [M−1 cm−1]) = 644 (6000), 589 (13[thin space (1/6-em)]000), 511 (41[thin space (1/6-em)]000), 416 nm (231[thin space (1/6-em)]000).

{5,10,15-Tris(3-hydroxyphenyl)-20-[4-(((1-(3-hydroxypropyl)-1H-1,2,3-triazol-4-yl)methyl)amino)tetrafluorophenyl]porphyrinato}-zinc(II) (10a). In a 25 mL flask with magnetic stirrer {5,10,15-tris(3-hydroxyphenyl)-20-[4-(prop-2-yn-1-ylamino)tetrafluorophenyl]porphyrinato}-zinc(II) (2h) (43.4 mg, 51.0 μmol) was dissolved in 1 mL of anhydrous DMSO (Acros) under argon. To the stirred solution 3-azidopropanol (823 mg, 8.14 mmol), L-ascorbic acid sodium salt (20.4 μL, 0.50 M in H2O, 10.2 μmol), and copper(II) sulfate pentahydrate (12.8 μL, 0.40 M in H2O, 5.10 μmol) were added. The solution was stirred for 30 min at RT. The crude product was diluted with 100 mL of ethyl acetate and was washed once with 100 mL of saturated NaCl solution. The aqueous layer was extracted three times with 50 mL of ethyl acetate. The combined organic layers were washed four times with 100 mL of saturated NaCl solution. Afterwards the organic layer was dried over Na2SO4. The crude product was evaporated to dryness and the remaining residue was purified by column chromatography (DCM/methanol = 95/5, v/v, Fluka) and recrystallization from DCM to obtain {5,10,15-tris(3-hydroxyphenyl)-20-[4-(((1-(3-hydroxypropyl)-1H-1,2,3-triazol-4-yl)methyl)amino)tetrafluorophenyl]porphyrinato}-zinc(II) (10a) (47.7 mg, 45.4 μmol, 89% yield) as purple-red solid.

1H NMR (THF-D8, 700 MHz): δ = 8.97 (d, 3J(H,H) = 4.5 Hz, 2H, 2,18-β), 8.92 (d, 3J(H,H) = 4.5 Hz, 2H, 7,13-β), 8.90 (d, 3J(H,H) = 4.5 Hz, 2H, 8,12-β), 8.88 (d, 3J(H,H) = 4.5 Hz, 2H, 3,17-β), 8.84 (s, 2H, 5,15-meso-Ar-OH), 8.83 (s, 1H, 10-meso-Ar-OH), 7.95 (s, 1H, triazole-H), 7.65–7.62 (m, 6H, 5,10,15-meso-2,6-Ar), 7.508 (t, 3J(H,H) = 8.1 Hz, 2H, 5,15-meso-5-Ar), 7.506 (t, 3J(H,H) = 8.1 Hz, 1H, 10-meso-5-Ar), 7.184 (dt, 3J(H,H) = 8.4 Hz, 4J(H,H) = 1.1 Hz, 2H, 5,15-meso-4-Ar), 7.181 (dt, 3J(H,H) = 8.4 Hz, 4J(H,H) = 1.1 Hz, 1H, 10-meso-4-Ar), 6.15 (t, 3J(H,H) = 6.7 Hz, 1H, ArF-NH), 4.89 (d, 3J(H,H) = 6.8 Hz, 2H, ArF-NHCH2), 4.55 (t, 3J(H,H) = 7.1 Hz, 2H, triazole-NCH2), 3.98 (t, 3J(H,H) = 5.0 Hz, 1H, CH2OH), 2.14–2.09 ppm (m, 2H, CH2CH2OH). 13C NMR (THF-D8, 176 MHz): δ = 157.02, 157.00, 151.28, 150.96, 150.78, 148.68, 147.32, 146.52, 145.57, 145.51, 139.01, 137.66, 133.33, 132.46, 132.18, 130.62, 129.57, 127.85, 127.81, 127.14, 127.12, 127.09, 127.06, 127.02, 123.20, 123.17, 123.13, 122.99, 122.84, 121.87, 115.27, 110.29, 103.47, 59.05, 47.73, 42.00, 34.41 ppm. 19F NMR (THF-D8, 376 MHz): δ = −142.62–(−142.92) (m, 2F, m-ArF), −162.03 ppm (d, 3J(F,F) = 17.6 Hz, 2F, o-ArF). m.p.: >300 °C. HRMS (ESI): calc. for C50H35F4N8O4Zn+ ([M + H]+): 951.2009; found: 951.1966.

{5,10,15-Tris(3-hydroxyphenyl)-20-[4-(((1-(3-azidopropyl)-1H-1,2,3-triazol-4-yl)methyl)amino)tetrafluorophenyl]porphyrinato}-zinc(II) (10b). In a 25 mL flask with magnetic stirrer {5,10,15-tris(3-hydroxyphenyl)-20-[4-(prop-2-yn-1-ylamino)tetrafluorophenyl]porphyrinato}-zinc(II) (2h) (103 mg, 121 μmol) was dissolved in 4 mL of anhydrous DMSO (Acros) under argon. To the stirred solution 1,3-diazidopropane (1.60 g, 12.7 mmol), L-ascorbic acid sodium salt (≥99%, 75.0 mg, 375 μmol), and copper(II) sulfate pentahydrate (32.0 mg, 128 μmol) were added. The solution was stirred for 30 min at RT. The crude product was diluted with 100 mL of ethyl acetate and was washed once with 100 mL of saturated NaCl solution. The aqueous layer was extracted three times with 50 mL of ethyl acetate. The combined organic layers were washed four times with 100 mL of saturated NaCl solution. Afterwards the organic layer was dried over Na2SO4. The crude product was evaporated to dryness and the remaining residue was purified by column chromatography (DCM/methanol = 96/4, v/v → 85/15, v/v, Fluka) to obtain two fractions. Both fractions were recrystallized from n-pentane to obtain: fraction 1 {5,10,15-tris(3-hydroxyphenyl)-20-[4-(((1-(3-azidopropyl)-1H-1,2,3-triazol-4-yl)methyl)amino)tetrafluorophenyl]porphyrinato}-zinc(II) (10b) (43.4 mg, 44.4 μmol, 37% yield) and fraction 2 porphyrin-dimer (10c) (44.2 mg, 24.2 μmol, 40% yield) as purple-red solids.
Porphyrin 10b. 1H NMR (acetone-D6, 700 MHz): δ = 8.98 (d, 3J(H,H) = 4.5 Hz, 2H, 2,18-β), 8.97–8.94 (m, 6H, 3,7,8,12,13,17-β), 8.75–8.70 (bs, 3H, 5,10,15-meso-3-Ar-OH), 7.90 (s, 1H, triazole-H), 7.74–7.72 (m, 3H, 5,10,15-meso-2-Ar), 7.72–7.69 (m, 3H, 5,10,15-meso-6-Ar), 7.59 (t, 3J(H,H) = 7.8 Hz, 3H, 5,10,15-meso-5-Ar), 7.29 (dd, 3J(H,H) = 8.4 Hz, 4J(H,H) = 2.4 Hz, 3H, 5,10,15-meso-4-Ar), 5.63 (t, 3J(H,H) = 7.3 Hz, 1H, ArF-NH), 4.39 (t, 3J(H,H) = 6.8 Hz, 2H, triazole-NCH2), 4.32 (d, 3J(H,H) = 7.6 Hz, 2H, ArF-NHCH2), 3.29 (t, 3J(H,H) = 6.6 Hz, 2H, N3CH2), 2.09 ppm (t, 3J(H,H) = 6.7 Hz, 2H, N3CH2CH2). 13C NMR (acetone-D6, 176 MHz): δ = 156.52, 156.50, 151.16, 151.11, 150.85, 150.68, 148.41, 147.06, 146.24, 145.38, 145.30, 138.98, 137.62, 133.47, 132.69, 132.39, 130.98, 129.18, 128.23, 128.20, 127.29, 123.14, 122.92, 122.77, 121.78, 115.41, 115.34, 110.37, 103.55, 48.95, 47.84, 41.28, 41.20 ppm. 19F NMR (acetone-D6, 471 MHz): δ = −142.70 (d, 3J(F,F) = 21.7 Hz, 2F, m-ArF), −161.13–(−161.51) ppm (m, 2F, o-ArF). m.p.: >300 °C. HRMS (ESI): calc. for C50H32F4N11O3Zn ([M − H]): 974.1922; found: 974.2182. UV/Vis (DCM): λmax (ε [M−1 cm−1]) = 647 (4000), 595 (4000), 553 (19[thin space (1/6-em)]000), 515 (20[thin space (1/6-em)]000), 422 nm (20[thin space (1/6-em)]000).
Porphyrin dimer 10c. 1H NMR (acetone-D6, 700 MHz): δ = 8.97–8.93 (m, 12H, 3,7,8,12,13,17-β), 8.91 (d, 3J(H,H) = 4.4 Hz, 2H, 2,18-β), 8.81–8.74 (m, 6H, 5,10,15-meso-3-Ar-OH), 7.74 (m, 6H, 5,10,15-meso-2-Ar), 7.71–7.66 (m, 6H, 5,10,15-meso-6-Ar), 7.58–7.51 (m, 8H, 5,10,15-meso-5-Ar + triazole-H), 7.29–7.24 (m, 6H, 5,10,15-meso-4-Ar), 5.23–5.15 (bs, 2H, ArF-NH), 3.91–3.82 (bs, 4H, triazole-NCH2), 3.70–3.60 (bs, 4H, ArF-NHCH2), 2.04–1.99 ppm (m, 2H, triazole-NCH2CH2). 13C NMR (acetone-D6, 176 MHz): δ = 156.52, 156.49, 156.41, 151.17, 151.09, 150.87, 150.69, 148.33, 146.97, 145.71, 145.43, 145.32, 138.82, 137.47, 133.49, 132.67, 132.38, 131.00, 128.85, 128.20, 128.16, 127.30, 123.15, 122.99, 122.92, 122.77, 121.80, 115.40, 110.52, 103.46, 47.42, 40.58 ppm. 19F NMR (acetone-D6, 376 MHz): δ = −141.69–(−142.99) (m, 4F, m-ArF), −161.08 ppm (d, 3J(F,F) = 16.3 Hz, 4F, o-ArF). m.p.: >300 °C. HRMS (ESI): calc. for C97H61F8N16O6Zn2+ ([M + H]+): 1825.3410; found: 1827.3568. UV/Vis (methanol): λmax (ε [M−1 cm−1]) = 647 (8000), 594 (9000), 553 (36[thin space (1/6-em)]000), 515 (35[thin space (1/6-em)]000), 422 nm (36[thin space (1/6-em)]000).
{5,10,15-Tris(3-hydroxyphenyl)-20-[4-(((1-(3-(4-(((mannosyl-6-(hydroxymethyl)tetrahydro-2H-pyran-2-yl)oxy)methyl)-1H-1,2,3-triazol-1-yl)propyl)-1H-1,2,3-triazol-4-yl)methyl)amino)tetrafluorophenyl]porphyrinato}-zinc(II) (10d). In a 25 mL flask with magnetic stirrer {5,10,15-tris(3-hydroxyphenyl)-20-[4-(((1-(3-azidopropyl)-1H-1,2,3-triazol-4-yl)methyl)amino)tetrafluorophenyl]porphyrinato}-zinc(II) (10b) (20.8 mg, 21.3 μmol) was dissolved in 3 mL of anhydrous DMSO (Acros) under argon. To the stirred solution propargyl-α-D-mannopyranoside (8.60 mg, 39.4 μmol), L-ascorbic acid sodium salt (≥99%, 15.0 mg, 75.0 μmol), and copper(II) sulfate pentahydrate (5.00 mg, 20.0 μmol) were added. The solution was stirred for 1 h at RT. The crude product was diluted with 100 mL of ethyl acetate and was washed once with 100 mL of saturated NaCl solution. The aqueous layer was extracted three times with 50 mL of ethyl acetate. The combined organic layers were washed four times with 100 mL of saturated NaCl solution. Afterwards the organic layer was dried over Na2SO4. The crude product was evaporated to dryness and the remaining residue was purified by column chromatography (DCM/methanol = 85/15, v/v, Fluka) and recrystallization from DCM to obtain {5,10,15-tris(3-hydroxyphenyl)-20-[4-(((1-(3-(4-(((mannosyl-6-(hydroxymethyl)tetrahydro-2H-pyran-2-yl)oxy)methyl)-1H-1,2,3-triazol-1-yl)propyl)-1H-1,2,3-triazol-4-yl)methyl)amino)tetrafluorophenyl]porphyrinato}-zinc(II) (10d) (24.1 mg, 20.2 μmol, 95% yield) as a purple-red solid.

1H NMR (CD3OD, 500 MHz): δ = 8.94 (d, 3J(H,H) = 4.7 Hz, 2H, 2,18-β), 8.93–8.87 (m, 4H, 7,8,12,13-β), 8.83 (d, 3J(H,H) = 5.0 Hz, 2H, 3,17-OH), 8.01 (s, 1H, NHCH2-triazole-H), 7.92 (s, 1H, OCH2-triazole-H), 7.71–7.63 (m, 6H, 5,10,15-meso-2,6-Ar), 7.59–7.51 (m, 3H, 5,10,15-meso-5-Ar), 7.26–7.19 (m, 3H, 5,10,15-meso-4-Ar), 4.84–4.79 (m, 2H, ArF-NHCH2), 4.71 (d, 2J(H,H) = 12.4 Hz, 1H, OCH2-triazole), 4.58 (s, 1H, Man-H-1), 4.54 (d, 2J(H,H) = 12.3 Hz, 1H, OCH2-triazole), 4.44 (t, 3J(H,H) = 6.8 Hz, 2H, ArF-NHCH2-triazole-CH2), 4.40 (t, 3J(H,H) = 6.9 Hz, 2H, OCH2-triazole-CH2), 3.81 (dd, 2J(H,H) = 12.1 Hz, 3J(H,H) = 3.2 Hz, 1H, Man-H-6b), 3.75 (dd, 3J(H,H) = 3.6 Hz, 3J(H,H) = 1.9 Hz, 1H, Man-H-2), 3.69 (dd, 2J(H,H) = 11.8 Hz, 3J(H,H) = 5.9 Hz, 1H, Man-H-6a), 3.68–3.61 (m, 1H, Man-H-3), 3.58 (t, 3J(H,H) = 9.4 Hz, 1H, Man-H-4), 3.58–3.46 (m, 1H, Man-H-5), 2.51 ppm (t, 3J(H,H) = 6.9 Hz, 2H, ArF-NHCH2-triazole-CH2CH2). 13C NMR (CD3OD, 126 MHz): δ = 156.69, 151.69, 151.60, 151.34, 151.17, 147.96, 145.93, 145.48, 133.65, 132.76, 132.47, 130.83, 128.31, 127.77, 125.56, 124.45, 123.27, 122.11, 115.44, 103.65, 100.81, 74.90, 72.47, 71.98, 71.13, 68.61, 62.95, 60.72, 41.71, 31.59, 30.72, 30.49 ppm. 19F NMR (CD3OD, 376 MHz): δ = −143.22 (d, 3J(F,F) = 21.5 Hz, 2F, m-ArF), −162.12 ppm (d, 3J(F,F) = 20.1 Hz, 2F, o-ArF). m.p.: 225 °C. HRMS (ESI): calc. for C59H47F4N11O9NaZn+ ([M + Na]+): 1216, 2678; found: 1216, 2535. UV/Vis (methanol): λmax (ε [M−1 cm−1]) = 647 (6000), 595 (4000), 555 (4000), 515 (25[thin space (1/6-em)]000), 422 nm (23[thin space (1/6-em)]000).

{5,10,15-Tris(3-benzyloxyphenyl)-5-[2,3,5,6-tetrafluoro-4-((1-((2R,3R,4S,5R,6R)-3,4,5-triacetoxy-6-(acetoxymethyl)tetrahydro-2H-pyran-2-yl)-1H-1,2,3-triazol-4-yl)methylamino)phenyl]porphyrinato}-zinc(II) (11). In a 25 mL flask with magnetic stirrer acetobromo-alpha-D-glucose (98%, 110 mg, 263 μmol) was dissolved in 3.4 mL of anhydrous DMSO (Roth). NaN3 (99%, 21.0 mg, 320 μmol) was added and the mixture was stirred for 10 min at RT. {5,10,15-tris(3-benzyloxyphenyl)-20-[4-(prop-2-ynylamino)tetrafluorophenyl]porphyrinato}-zinc(II) (2i) (150 mg, 134 μmol), L-ascorbic acid sodium salt (700 μL, 1.43 M in H2O, 1.00 mmol), and copper(II) sulfate pentahydrate (700 μL, 1.43 M in H2O, 1.00 mmol) were added and the solution was stirred at RT for 52 h. Portions of the reactants were added after 16 h (acetobromo-alpha-D-glucose (98%, 110 mg, 263 μmol) and NaN3 (99%, 21.0 mg, 320 μmol) dissolved in 2 mL of anhydrous DMSO (Roth), L-ascorbic acid sodium salt (700 μL, 1.43 M in H2O, 1.00 mmol) and copper(II) sulfate pentahydrate (700 μL, 1.43 M in H2O, 1.00 mmol)), 32 h (acetobromo-alpha-D-glucose (98%, 550 mg, 1.31 mmol) and NaN3 (99%, 105 mg, 1.60 mmol) dissolved in 10 mL of anhydrous DMSO (Roth), L-ascorbic acid sodium salt (3.30 mL, 1.52 M in H2O, 5.00 mmol) and copper(II) sulfate pentahydrate (3.30 μL, 1.52 M in H2O, 5.01 mmol)), and 48 h (acetobromo-alpha-D-glucose (98%, 275 mg, 656 μmol) and NaN3 (99%, 52.5 mg, 800 μmol) dissolved in 5 mL of anhydrous DMSO (Roth), L-ascorbic acid sodium salt (1.70 mL, 1.47 M in H2O, 2.50 mmol) and copper(II) sulfate pentahydrate (1.70 μL, 1.47 M in H2O, 2.50 mmol)) of stirring. Three drops DIPEA were added and the reaction mixture was stirred for 1 h. The crude product was diluted with 100 mL of DCM and washed three times with 50 mL of H2O. Afterwards the organic layer was dried over Na2SO4. The crude product was evaporated to dryness and the remaining residue was purified by column chromatography (DCM/ethyl acetate = 9/1, v/v, Machery-Nagel) and recrystallization from DCM/methanol to obtain {5,10,15-tris(3-benzyloxyphenyl)-5-[2,3,5,6-tetrafluoro-4-((1-((2R,3R,4S,5R,6R)-3,4,5-triacetoxy-6-(acetoxymethyl)tetrahydro-2H-pyran-2-yl)-1H-1,2,3-triazol-4-yl)methylamino)phenyl]porphyrinato}-zinc(II) (11) (34.0 mg, 22.7 μmol, 17% yield) as a pink solid.

1H NMR (CDCl3, 500 MHz): δ = 9.01–8.93 (m, 6H, 3,7,8,12,13,17-β), 8.88 (d, 3J(H,H) = 4.3 Hz, 2H, 2,18-β), 7.89–7.80 (m, 6H, Ar), 7.66–7.58 (m, 3H, Ar), 7.43 (s, 1H, triazole-H), 7.39–7.10 (m, 18H, Ar), 5.55 (d, 3J(H,H) = 9.2 Hz, 1H, H-1 ose), 5.34–5.27 (m, 1H, H-3 ose), 5.21 (t, 3J(H,H) = 9.4 Hz, 1H, H-4 ose), 5.17–5.01 (m, 9H, CH2 + H-2 ose), 4.19 (dd, vicinal: 3J(H,H) = 12.7 Hz, geminal: 2J(H,H) = 4.7 Hz, 1H, H-6 ose), 4.06–3.99 (m, 1H, H-5 ose), 3.90–3.83 (m, 1H, H-6 ose), 2.02 (s, 3H, OAc), 1.95 (s, 3H, OAc), 1.92 (s, 3H, OAc), 1.65 ppm (s, 3H, OAc). 13C NMR (CDCl3, 126 MHz): δ = 170.57, 169.95, 169.40, 168.85, 157.06, 150.55, 150.31, 150.19, 150.03, 144.22, 144.17, 136.92, 136.84, 133.04, 132.41, 132.12, 130.65, 128.57, 128.07, 128.01, 127.94, 127.66, 127.59, 127.55, 127.52, 122.14, 121.47, 121.42, 121.15, 119.98, 114.63, 114.59, 111.10, 85.85, 75.26, 72.33, 70.28, 70.20, 67.67, 61.44, 20.71, 20.63, 20.58, 19.95 ppm. 19F NMR (CDCl3, 471 MHz): δ = −140.23–(−140.53) (m, 2F, m-ArF), −159.31–(−159.55) ppm (m, 2F, o-ArF). m.p.: 120 °C. HRMS (ESI): calc. for C82H64F4N8O12Zn+ ([M]+): 1492.3871 found: 1492.3994. UV/Vis (DCM): λmax (ε [M−1 cm−1]) = 585 (3000), 548 (17[thin space (1/6-em)]000), 513 (19[thin space (1/6-em)]000), 420 nm (260[thin space (1/6-em)]000).

Porphyrin-hPG19.5-conjugate with 3% porphyrins and 10% azides 13a. In a 10 mL flask with magnetic stirrer hPG19.5-azide with 13% azides 12a (68.0 mg, 3.34 μmol, 114 μmol azido groups) was dissolved in 1 mL of anhydrous DMSO (Acros). {5,10,15-Tris(3-hydroxyphenyl)-20-[4-(prop-2-yn-1-ylamino)tetrafluorophenyl]porphyrinato}-zinc(II) (2h) (22.0 mg, 26.0 μmol), L-ascorbic acid sodium salt (26.0 μL, 0.5 M in H2O, 13.0 μmol), and copper(II) sulfate pentahydrate (16.0 μL, 0.40 M in H2O, 6.40 μmol) were added and the solution was stirred at RT for 2 d. The crude product was purified by dialysis (acetone/H2O = 9/1, v/v) for 2 d to obtain the purple wax-like product porphyrin-hPG19.5-conjugate with 3% porphyrins and 10% azides 13a (75.0 mg, 2.89 μmol, 19.0 μmol porphyrin and 80.0 μmol azido groups, 87% yield, 84% conversion).

1H NMR (acetone-D6/D2O = 5/1, v/v, 700 MHz): δ = 9.16–8.28 (bs, β), 7.86–6.53 (m, Ar + triazole-H), 4.05–2.72 ppm (m, hPG-backbone + porphyrin-CH2). 13C NMR (acetone-D6/D2O = 5/1, v/v, 176 MHz): δ = 155.71, 150.64, 150.37, 150.13, 144.77, 133.14, 132.25, 131.98, 130.53, 127.92, 126.85, 122.39, 121.30, 114.98, 80.50, 80.24, 78.95, 78.66, 72.94, 71.80, 71.36, 71.15, 71.12, 69.85, 69.55, 63.34, 61.53, 53.83 ppm. UV/Vis (acetone/H2O = 9/1, v/v): λmax = 598, 557, 424 nm. Mw,NMR = 26.000.

Porphyrin-hPG116-conjugate with 4% porphyrins and 1% azides 13b. In a 5 mL flask with magnetic stirrer hPG116-azide with 5% azides 12b (55.0 mg, 466 nmol, 36.5 μmol azido groups) was dissolved in 1 mL of anhydrous DMSO (Acros). {5,10,15-Tris(3-hydroxyphenyl)-20-[4-(prop-2-yn-1-ylamino)tetrafluorophenyl]porphyrinato}-zinc(II) (2h) (30.2 mg, 35.5 μmol), L-ascorbic acid sodium salt (252 μL, 26 mM in H2O, 6.55 μmol), and copper(II) sulfate pentahydrate (52.0 μL, 0.14 M in H2O, 7.05 μmol) were added and the solution was stirred at RT for 3 d. Afterwards the reaction mixture was heated to 40 °C for 3 h. The crude product was purified by dialysis (acetone/H2O = 4/1, v/v) for 6 d to obtain the purple wax-like product porphyrin-hPG116-conjugate with 4% porphyrins and 1% azides 13b (58.4 mg, 330 nmol, 22.8 μmol porphyrin and 3.10 μmol azido groups, 71% yield, 91% conversion).

1H NMR (D2O, 700 MHz): δ = 9.77–8.51 (bs, β), 8.51–6.98 (m, Ar + triazole-H), 4.32–2.62 ppm (m, hPG-backbone + porphyrin-CH2). 13C NMR (D2O, 176 MHz): δ = 154.80, 149.85, 145.88, 143.88, 136.69, 132.44, 127.70, 122.16, 115.05, 107.93, 79.42, 77.91, 72.12, 70.85, 70.69, 70.41, 69.16, 68.87, 62.60, 60.76 ppm. UV/Vis (H2O): λmax = 597, 557, 423 nm. Mw,NMR = 177.000.

Porphyrin-hPG19.5-conjugate with 0.4% porphyrins and 1.6% azides 13c. In a 10 mL flask with magnetic stirrer hPG19.5-azide with 2% azides 12c (56.0 mg, 2.85 μmol, 14.0 μmol azido groups) was dissolved in 1 mL of anhydrous DMSO (Acros). {5,10,15-Tris(3-hydroxyphenyl)-20-[4-(prop-2-yn-1-ylamino)tetrafluorophenyl]porphyrinato}-zinc(II) (2h) (5.0 mg, 7.05 μmol), L-ascorbic acid sodium salt (26.0 μL, 0.5 M in H2O, 13.0 μmol), and copper(II) sulfate pentahydrate (16.0 μL, 0.40 M in H2O, 6.40 μmol) were added and the solution was stirred at RT for 5 min. The crude product was purified by dialysis (methanol/H2O = 4/1, v/v) for 2 d to obtain the purple product porphyrin-hPG19.5-conjugate 0.4% porphyrins and 1.6% azides 13c. The product was directly converted to 14a in the next reaction without drying.
Porphyrin-mPEG-hPG19.5-conjugate with 0.4% porphyrins, 1.3% mPEG, and 0.3% azides 14a. In a 10 mL flask with magnetic stirrer porphyrin-hPG19.5-conjugate 0.4% porphyrins and 1.6% azides 13c was dissolved in 3 mL of H2O. mPEG propargyl ether (average MW = 350) (7.0 mg, 20.0 μmol), L-ascorbic acid sodium salt (26.0 μL, 0.5 M in H2O, 13.0 μmol), and copper(II) sulfate pentahydrate (16.0 μL, 0.40 M in H2O, 6.40 μmol) were added and the solution was stirred at RT for 1 d. The crude product was purified by dialysis (H2O) for 2 d to obtain the purple wax-like product porphyrin-mPEG-hPG19.5-conjugate with 0.4% porphyrins, 1.3% mPEG, and 0.3% azides 14a (53.0 mg, 2.38 μmol, 1.05 μmol porphyrin, 3.43 μmol mPEG, and 791 nmol azido groups, 84% yield over two steps).

1H NMR (D2O, 700 MHz): δ = 9.22–8.63 (m, β), 8.42–7.15 (m, Ar + triazole-H), 4.32–3.35 (m, hPG-backbone + porphyrin-CH2 + mPEG-CH3), 1.38 (s, CH2-hPG starter unit), 0.89 ppm (CH3-hPG starter unit). 13C NMR (D2O, 176 MHz): δ = 79.63, 79.41, 78.14, 77.90, 72.12, 70.97, 70.86, 70.69, 70.40, 69.56, 69.42, 69.16, 68.86, 62.59, 60.73, 58.04 ppm. UV/Vis (H2O): λmax = 600, 559, 429 nm. Mw,NMR = 22.300.

Porphyrin-mPEG-hPG19.5-conjugate with 3% porphyrins, 3% mPEG, and 7% azides 14b. In a 10 mL flask with magnetic stirrer porphyrin-hPG19.5-conjugate 3% porphyrins and 10% azides 13a (59.0 mg, 2.11 μmol, 18.9 μmol porphyrin and 53 μmol azido groups) was dissolved in 2.2 mL of acetone and 800 μL of H2O. mPEG propargyl ether (average MW = 350) (22.0 mg, 62.9 μmol), L-ascorbic acid sodium salt (26.0 μL, 0.5 M in H2O, 13.0 μmol), and copper(II) sulfate pentahydrate (21.0 μL, 0.30 M in H2O, 6.30 μmol) were added and the solution was stirred at RT for 2 d. The crude product was purified by dialysis (acetone/H2O = 4/1, v/v) for 2 d to obtain the purple wax-like product porphyrin-mPEG-hPG19.5-conjugate with 3% porphyrins, 3% mPEG and 7% azides 14b (62.0 mg, 1.99 μmol, 17.8 μmol porphyrin, 17.8 μmol mPEG and 32.5 μmol azido groups, 94% yield, 35% conversion).

1H NMR (acetone-D6/D2O = 4/1, v/v, 700 MHz): δ = 9.23–8.44 (m, β), 8.20–6.85 (m, Ar + triazole-H), 4.17–2.50 (m, hPG-backbone + porphyrin-CH2 + mPEG-CH3), 1.31 (s, CH2-hPG starter unit), 0.80 ppm (CH3-hPG starter unit). 13C NMR (acetone-D6/D2O = 4/1, v/v, 176 MHz): δ = 150.02, 144.62, 94.49, 80.19, 79.98, 78.69, 78.44, 72.69, 71.50, 71.16, 70.91, 70.08, 69.64, 69.35, 63.10, 61.30, 58.39, 53.58, 51.50 ppm. UV/Vis (acetone-D6/D2O = 4/1, v/v): λmax = 597, 556, 423 nm. Mw,NMR = 31.200.

Acknowledgements

This work was supported by the SFB 765 funded by the German Science Foundation (DFG). We thank our colleagues Florian Paulus, Julian Heinrich and Joseph Schutt for their scientific support.

Notes and references

  1. D. E. J. G. J. Dolmans, D. Fukumura and R. K. Jain, Nat. Rev. Cancer, 2003, 3, 380–387 CrossRef CAS PubMed.
  2. M. Ethirajan, Y. Chen, P. Joshi and R. K. Pandey, Chem. Soc. Rev., 2011, 40, 340–362 RSC.
  3. A. E. O'Connor, W. M. Gallagher and A. T. Byrne, Photochem. Photobiol., 2009, 85, 1053–1074 CrossRef PubMed.
  4. A. Ormond and H. Freeman, Materials, 2013, 6, 817 CrossRef CAS.
  5. J. Králová, T. Bříza, I. Moserová, B. Dolenský, P. Vašek, P. Poučková, Z. Kejík, R. Kaplánek, P. Martásek, M. Dvořák and V. Král, J. Med. Chem., 2008, 51, 5964–5973 CrossRef PubMed.
  6. L. B. Josefsen and R. W. Boyle, Theranostics, 2012, 2, 916–966 CrossRef CAS PubMed.
  7. D. Gust, T. A. Moore and A. L. Moore, Acc. Chem. Res., 2009, 42, 1890–1898 CrossRef CAS PubMed.
  8. L.-L. Li and E. W.-G. Diau, Chem. Soc. Rev., 2013, 42, 291–304 RSC.
  9. I. Aviv and Z. Gross, Chem. Commun., 2007, 1987–1999 RSC.
  10. E. Rose, B. Andrioletti, S. Zrig and M. Quelquejeu-Etheve, Chem. Soc. Rev., 2005, 34, 573–583 RSC.
  11. R. H. Morris, Chem. Soc. Rev., 2009, 38, 2282–2291 RSC.
  12. D. Kessel and C. J. Dutton, Photochem. Photobiol., 1984, 40, 403–405 CrossRef CAS PubMed.
  13. L. B. Rocha, L. C. Gomes-da-Silva, J. M. Dąbrowski and L. G. Arnaut, Eur. J. Cancer, 2015, 51, 1822–1830 CrossRef CAS PubMed.
  14. A. Aggarwal, S. Thompson, S. Singh, B. Newton, A. Moore, R. Gao, X. Gu, S. Mukherjee and C. M. Drain, Photochem. Photobiol., 2014, 90, 419–430 CrossRef CAS PubMed.
  15. A. Hajri, S. Wack, C. Meyer, M. K. Smith, C. Leberquier, M. Kedinger and M. Aprahamian, Photochem. Photobiol., 2002, 75, 140–148 CrossRef CAS PubMed.
  16. S. Singh, A. Aggarwal, S. Thompson, J. P. C. Tome, X. C. Zhu, D. Samaroo, M. Vinodu, R. M. Gao and C. M. Drain, Bioconjugate Chem., 2010, 21, 2136–2146 CrossRef CAS PubMed.
  17. R. Bonnett, R. D. White, U. J. Winfield and M. C. Berenbaum, Biochem. J., 1989, 261, 277–280 CrossRef CAS PubMed.
  18. E. Ben-Hur and I. Rosenthal, Int. J. Radiat. Biol., 1985, 47, 145–147 CAS.
  19. L. M. O. Lourenco, P. M. R. Pereira, E. Maciel, M. Valega, F. M. J. Domingues, M. R. M. Domingues, M. Neves, J. A. S. Cavaleiro, R. Fernandes and J. P. C. Tome, Chem. Commun., 2014, 50, 8363–8366 RSC.
  20. D. Aviezer, S. Cotton, M. David, A. Segev, N. Khaselev, N. Galili, Z. Gross and A. Yayon, Cancer Res., 2000, 60, 2973–2980 CAS.
  21. J. F. B. Barata, A. Zamarrón, M. G. P. M. S. Neves, M. A. F. Faustino, A. C. Tomé, J. A. S. Cavaleiro, B. Röder, Á. Juarranz and F. Sanz-Rodríguez, Eur. J. Med. Chem., 2015, 92, 135–144 CrossRef CAS PubMed.
  22. K. M. Kadish, C. Araullomcadams, B. C. Han and M. M. Franzen, J. Am. Chem. Soc., 1990, 112, 8364–8368 CrossRef CAS.
  23. N. V. S. D. K. Bhupathiraju, W. Rizvi, J. D. Batteas and C. M. Drain, Org. Biomol. Chem., 2016, 14, 389–408 CAS.
  24. T. Goslinski and J. Piskorz, J. Photochem. Photobiol., C, 2011, 12, 304–321 CrossRef CAS.
  25. C. S. Gutsche, M. Ortwerth, S. Gräfe, K. J. Flanagan, M. O. Senge, H.-U. Reissig, N. Kulak and A. Wiehe, Chem. – Eur. J., 2016 DOI:10.1002/chem.201601857.
  26. P. Battioni, O. Brigaud, H. Desvaux, D. Mansuy and T. G. Traylor, Tetrahedron Lett., 1991, 32, 2893–2896 CrossRef CAS.
  27. H. R. A. Golf, H.-U. Reissig and A. Wiehe, Eur. J. Org. Chem., 2015, 1548–1568 CrossRef CAS.
  28. H. R. A. Golf, H.-U. Reissig and A. Wiehe, Org. Lett., 2015, 17, 982–985 CrossRef CAS PubMed.
  29. V. A. Olshevskaya, A. V. Zaitsev, A. L. Sigan, E. G. Kononova, P. V. Petrovskii, N. D. Chkanikov and V. N. Kalinin, Dokl. Chem., 2010, 435, 334–338 CrossRef CAS.
  30. R. Weiss, F. Puhlhofer, N. Jux and K. Merz, Angew. Chem., Int. Ed., 2002, 41, 3815–3817 CrossRef CAS.
  31. C. F. Pereira, J. A. Fernandes, J. M. M. Rodrigues, S. M. F. Vilela, J. P. C. Tome and F. A. Almeida Paz, Acta Crystallogr., Sect. C: Cryst. Struct. Commun., 2012, 68, O104–O107 CAS.
  32. S. J. Shaw, K. J. Elgie, C. Edwards and R. W. Boyle, Tetrahedron Lett., 1999, 40, 1595–1596 CrossRef CAS.
  33. D. Steinhilber, M. Witting, X. Zhang, M. Staegemann, F. Paulus, W. Friess, S. Küchler and R. Haag, J. Controlled Release, 2013, 169, 289–295 CrossRef CAS PubMed.
  34. M. Calderón, M. A. Quadir, S. K. Sharma and R. Haag, Adv. Mater., 2010, 22, 190–218 CrossRef PubMed.
  35. R. K. Kainthan and D. E. Brooks, Biomaterials, 2007, 28, 4779–4787 CrossRef CAS PubMed.
  36. R. K. Kainthan, J. Janzen, E. Levin, D. V. Devine and D. E. Brooks, Biomacromolecules, 2006, 7, 703–709 CrossRef CAS PubMed.
  37. A. Sunder, R. Hanselmann, H. Frey and R. Mülhaupt, Macromolecules, 1999, 32, 4240–4246 CrossRef CAS.
  38. B. Jang and Y. Choi, Theranostics, 2012, 2, 190–197 CrossRef CAS PubMed.
  39. V. Cuchelkar, P. Kopečková and J. Kopeček, Macromol. Biosci., 2008, 8, 375–383 CrossRef CAS PubMed.
  40. C. Komeda, A. Ikeda, J.-i. Kikuchi, N. Ishida-Kitagawa, H. Tatebe, K. Shiozaki and M. Akiyama, Org. Biomol. Chem., 2013, 11, 2567–2570 CAS.
  41. T. Zhang, H. Lin, L. Cui, N. An, R. Tong, Y. Chen, C. Yang, X. Li, J. Liu and F. Qu, Eur. J. Inorg. Chem., 2016, 1206–1213 CrossRef CAS.
  42. D. K. Chatterjee, L. S. Fong and Y. Zhang, Adv. Drug Delivery Rev., 2008, 60, 1627–1637 CrossRef CAS PubMed.
  43. H.-L. Tu, Y.-S. Lin, H.-Y. Lin, Y. Hung, L.-W. Lo, Y.-F. Chen and C.-Y. Mou, Adv. Mater., 2009, 21, 172–177 CrossRef CAS.
  44. M. E. Wieder, D. C. Hone, M. J. Cook, M. M. Handsley, J. Gavrilovic and D. A. Russell, Photochem. Photobiol. Sci., 2006, 5, 727–734 CAS.
  45. R. Langer, Nature, 1998, 392, 5–10 CAS.
  46. K. E. Uhrich, S. M. Cannizzaro, R. S. Langer and K. M. Shakesheff, Chem. Rev., 1999, 99, 3181–3198 CrossRef CAS PubMed.
  47. H. Wei, R.-X. Zhuo and X.-Z. Zhang, Prog. Polym. Sci., 2013, 38, 503–535 CrossRef CAS.
  48. M. H. Staegemann, Master Thesis, Freie Universität, Berlin, 2011.
  49. M. O. Senge and J. C. Brandt, Photochem. Photobiol., 2011, 87, 1240–1296 CrossRef CAS PubMed.
  50. W. König and R. Geiger, Chem. Ber., 1970, 103, 788–798 CrossRef.
  51. G. Saito, J. A. Swanson and K.-D. Lee, Adv. Drug Delivery Rev., 2003, 55, 199–215 CrossRef CAS PubMed.
  52. N. Ballatori, S. M. Krance, S. Notenboom, S. Shujie, T. Kim and C. L. Hammond, Biol. Chem., 2009, 390, 191–214 CrossRef CAS PubMed.
  53. F. Q. Schafer and G. R. Buettner, Free Radical Biol. Med., 2001, 30, 1191–1212 CrossRef CAS PubMed.
  54. X. Zhang, K. Achazi, D. Steinhilber, F. Kratz, J. Dernedde and R. Haag, J. Controlled Release, 2014, 174, 209–216 CrossRef CAS PubMed.
  55. X. Zhang, S. Malhotra, M. Molina and R. Haag, Chem. Soc. Rev., 2015, 44, 1948–1973 RSC.
  56. I. Mellman, R. Fuchs and A. Helenius, Annu. Rev. Biochem., 1986, 55, 663–700 CrossRef CAS PubMed.
  57. H. R. A. Golf, PhD thesis, Freie Universität Berlin, 2015.
  58. H. R. A. Golf, A. Wiehe, S. Gräfe, V. Albrecht and H.-U. Reissig, US Pat., 62057353, 2015 Search PubMed.
  59. V. D. Bock, H. Hiemstra and J. H. van Maarseveen, Eur. J. Org. Chem., 2006, 51–68 CrossRef CAS.
  60. D. Fournier, R. Hoogenboom and U. S. Schubert, Chem. Soc. Rev., 2007, 36, 1369–1380 RSC.
  61. J.-F. Lutz, Angew. Chem., Int. Ed., 2007, 46, 1018–1025 CrossRef CAS PubMed.
  62. C. Hein, X.-M. Liu and D. Wang, Pharm. Res., 2008, 25, 2216–2230 CrossRef CAS PubMed.
  63. G. C. Tron, T. Pirali, R. A. Billington, P. L. Canonico, G. Sorba and A. A. Genazzani, Med. Res. Rev., 2008, 28, 278–308 CrossRef CAS PubMed.
  64. M. Claire, M. S. Eoin and M. O. Senge, Curr. Med. Chem., 2015, 22, 2238–2348 CrossRef.
  65. S. Singh, A. Aggarwal, N. V. S. D. K. Bhupathiraju, G. Arianna, K. Tiwari and C. M. Drain, Chem. Rev., 2015, 115, 10261–10306 CrossRef CAS PubMed.
  66. O. Warburg, Science, 1956, 123, 309–314 CAS.
  67. J. Yun, C. Rago, I. Cheong, R. Pagliarini, P. Angenendt, H. Rajagopalan, K. Schmidt, J. K. V. Willson, S. Markowitz, S. Zhou, L. A. Diaz, V. E. Velculescu, C. Lengauer, K. W. Kinzler, B. Vogelstein and N. Papadopoulos, Science, 2009, 325, 1555–1559 CrossRef CAS PubMed.
  68. R. E. Airley and A. Mobasheri, Chemotherapy, 2007, 53, 233–256 CrossRef CAS PubMed.
  69. R. Daly, G. Vaz, A. M. Davies, M. O. Senge and E. M. Scanlan, Chem. – Eur. J., 2012, 18, 14671–14679 CrossRef CAS PubMed.
  70. D. Kushwaha and V. K. Tiwari, J. Org. Chem., 2013, 78, 8184–8190 CrossRef CAS PubMed.
  71. N. Sharon, FEBS Lett., 1987, 217, 145–157 CrossRef CAS PubMed.
  72. R. Ikeda, F. Saito, M. Matsuo, K. Kurokawa, K. Sekimizu, M. Yamaguchi and S. Kawamoto, J. Bacteriol., 2007, 189, 4815–4826 CrossRef CAS PubMed.
  73. H. Furuya and R. Ikeda, Microbiol., 2009, 155, 2707–2713 CrossRef CAS PubMed.
  74. M. Weinhart, D. Gröger, S. Enders, J. Dernedde and R. Haag, Biomacromolecules, 2011, 12, 2502–2511 CrossRef CAS PubMed.
  75. E. Mohammadifar, A. Nemati Kharat and M. Adeli, J. Mater. Chem. B, 2015, 3, 3896–3921 RSC.
  76. A. A. Mendelson, Q. Guan, I. Chafeeva, G. A. da Roza, J. N. Kizhakkedathu and C. Du, Peritoneal Dial. Int., 2013, 33, 15–27 CrossRef CAS PubMed.
  77. C. Du, A. A. Mendelson, Q. Guan, R. Chapanian, I. Chafeeva, G. da Roza and J. N. Kizhakkedathu, Biomaterials, 2014, 35, 1378–1389 CrossRef CAS PubMed.
  78. S. Roller, H. Zhou and R. Haag, Mol. Diversity, 2005, 9, 305–316 CrossRef CAS PubMed.
  79. H. Zeng, C. Schlesener, O. Cromwell, M. Hellmund, R. Haag and Z. Guan, Biomacromolecules, 2015, 16, 3869–3877 CrossRef CAS PubMed.
  80. T. Pecchioli, M. K. Muthyala, R. Haag and M. Christmann, Beilstein J. Org. Chem., 2015, 11, 730–738 CrossRef CAS PubMed.
  81. R. Albrecht, S. Fehse, K. Pant, S. Nowag, H. Stephan, R. Haag and C. C. Tzschucke, Macromol. Biosci., 2016, 16, 412–419 CrossRef CAS PubMed.
  82. Y. Deng, J. K. Saucier-Sawyer, C. J. Hoimes, J. Zhang, Y.-E. Seo, J. W. Andrejecsk and W. M. Saltzman, Biomaterials, 2014, 35, 6595–6602 CrossRef CAS PubMed.
  83. M. Dzieciuch, S. Rissanen, N. Szydłowska, A. Bunker, M. Kumorek, D. Jamróz, I. Vattulainen, M. Nowakowska, T. Róg and M. Kepczynski, J. Phys. Chem. B, 2015, 119, 6646–6657 CrossRef CAS PubMed.
  84. J. M. Harris and R. B. Chess, Nat. Rev. Drug Discovery, 2003, 2, 214–221 CrossRef CAS PubMed.
  85. D. E. Owens Iii and N. A. Peppas, Int. J. Pharm., 2006, 307, 93–102 CrossRef PubMed.
  86. S. Barua and S. Mitragotri, Nano Today, 2014, 9, 223–243 CrossRef CAS PubMed.
  87. N. Bertrand, J. Wu, X. Xu, N. Kamaly and O. C. Farokhzad, Adv. Drug Delivery Rev., 2014, 66, 2–25 CrossRef CAS PubMed.
  88. J. Fang, H. Nakamura and H. Maeda, Adv. Drug Delivery Rev., 2011, 63, 136–151 CrossRef CAS PubMed.
  89. H. Maeda and Y. Matsumura, Crit. Rev. Ther. Drug Carrier Syst., 1989, 6, 193–210 CAS.
  90. C. A. Hunter, M. C. Misuraca and S. M. Turega, J. Am. Chem. Soc., 2011, 133, 582–594 CrossRef CAS PubMed.
  91. M. D. LoCoco, X. Zhang and R. F. Jordan, J. Am. Chem. Soc., 2004, 126, 15231–15244 CrossRef CAS PubMed.
  92. R. Haag and H. Türk, DE Pat., DE10211664A1, 2003 Search PubMed.
  93. A. Sunder, R. Mülhaupt, R. Haag and H. Frey, Adv. Mater., 2000, 12, 235–239 CrossRef CAS.
  94. I. N. Kurniasih, H. Liang, V. D. Moschwitzer, M. A. Quadir, M. Radowski, J. P. Rabe and R. Haag, New J. Chem., 2012, 36, 371–379 RSC.
  95. M. R. Patel, A. Bhatt, J. D. Steffen, A. Chergui, J. Murai, Y. Pommier, J. M. Pascal, L. D. Trombetta, F. R. Fronczek and T. T. Talele, J. Med. Chem., 2014, 57, 5579–5601 CrossRef CAS PubMed.
  96. T. Tanaka, B. S. Lee, N. Aratani, M.-C. Yoon, D. Kim and A. Osuka, Chem. – Eur. J., 2011, 17, 14400–14412 CrossRef CAS PubMed.
  97. P. J. Spellane, M. Gouterman, A. Antipas, S. Kim and Y. C. Liu, Inorg. Chem., 1980, 19, 386–391 CrossRef CAS.
  98. M. V. Berridge, P. M. Herst and A. S. Tan, in Biotechnol. Ann. Rev, Elsevier, 2005, vol. 11, pp. 127–152 Search PubMed.

Footnote

Electronic supplementary information (ESI) available. See DOI: 10.1039/c6ob01551d

This journal is © The Royal Society of Chemistry 2016