Next Article in Journal
Meloxicam and Study of Their Antimicrobial Effects against Phyto- and Human Pathogens
Next Article in Special Issue
Lipid Formulations and Bioconjugation Strategies for Indomethacin Therapeutic Advances
Previous Article in Journal
Chemical Composition of Tomato Seed Flours, and Their Radical Scavenging, Anti-Inflammatory and Gut Microbiota Modulating Properties
Previous Article in Special Issue
Synthesis and Biological Evaluation of 99mTc(I) Tricarbonyl Complexes Dual-Targeted at Tumoral Mitochondria
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Co-Opting Host Receptors for Targeted Delivery of Bioconjugates—From Drugs to Bugs

by
Kristen M. Tummillo
1,2 and
Karsten R.O. Hazlett
1,*
1
Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY 12208, USA
2
Admera Health, South Plainfield, NJ 07080, USA
*
Author to whom correspondence should be addressed.
Molecules 2021, 26(5), 1479; https://doi.org/10.3390/molecules26051479
Submission received: 1 January 2021 / Revised: 26 February 2021 / Accepted: 4 March 2021 / Published: 9 March 2021
(This article belongs to the Special Issue Bioconjugation Strategies in Drug Delivery and Molecular Imaging)

Abstract

:
Bioconjugation has allowed scientists to combine multiple functional elements into one biological or biochemical unit. This assembly can result in the production of constructs that are targeted to a specific site or cell type in order to enhance the response to, or activity of, the conjugated moiety. In the case of cancer treatments, selectively targeting chemotherapies to the cells of interest limit harmful side effects and enhance efficacy. Targeting through conjugation is also advantageous in delivering treatments to difficult-to-reach tissues, such as the brain or infections deep in the lung. Bacterial infections can be more selectively treated by conjugating antibiotics to microbe-specific entities; helping to avoid antibiotic resistance across commensal bacterial species. In the case of vaccine development, conjugation is used to enhance efficacy without compromising safety. In this work, we will review the previously mentioned areas in which bioconjugation has created new possibilities and advanced treatments.

1. Introduction

The process of conjugation has allowed scientists to improve therapies and vaccinations by linking therapeutic agents or protective antigens to targeting moieties that focus delivery of the cargo. Increasing the specificity of therapies promotes concentration of the agent which consequently reduces the needed systemic dose and minimizes side effects. Targeting of prophylactics and therapies can be achieved by fusing distinct functional moieties; a process often termed bioconjugation. Common methods of bioconjugation include genetic fusion, chemical conjugation, and in vivo bioconjugation; examples of each will be discussed in this review. Bioconjugation is broadly utilized from basic pre-clinical development to current clinical use and impacts medical disciplines from cancer, to neuropathies, to infectious disease.
In the case of treating cancer patients, systemic administration of chemotherapy regularly causes deleterious side effects and has limited efficacy; however, great strides have been made in limiting side effects and enhancing efficacy by specifically targeting therapies. Through targeted drug delivery, drugs can be concentrated in the desired tissue and specific anti-tumor cells can be activated [1,2,3,4,5]. The targeting concept has also been applied to neurodegenerative diseases with the goal of suppressing pathological immune responses and/or promoting repair pathways [6,7]. The ability to target antibiotics is a promising development as drug resistant pathogens have become increasingly prevalent and indiscriminate application of antibiotics promotes the development and spread of antibiotic resistance [8,9]. Further, conjugated vaccines have also been developed for many pathogenic microbes; the importance of the latter is glaringly highlighted by the COVID-19 pandemic.
The goal of this mini-review is to succulently highlight the breadth of receptor-targeting approaches employed by bioconjugate therapies and bioconjugate vaccines—both nascent and mature. To this end, we are skimming the surfaces of many broad fields (cancer, neuropathology, infectious disease) to show-case the diversity of creative targeting modalities.

2. Targeted Therapies

2.1. Cancer Therapies

Chemotherapy agents inhibit cell growth and division by inducing DNA damage, preventing DNA replication, and disrupting mitosis. Inhibiting the process of cell division is aimed at destruction of rapidly growing cancer cells, however, the division of rapidly dividing healthy cells is also susceptible to growth inhibition and killing by chemotherapy treatments. This leads to deleterious systemic side effects, such as hair loss and significant disruption of the gastrointestinal tract and immune system [10,11]. The severe side effects and poor therapeutic window(s) of conventional chemotherapy can leave patients weighing the risks and benefits of such treatments versus living with cancer. Therefore, there has been a great push for development of increased specificity and efficacy in cancer treatments. Conjugation of chemotherapy drugs to antibodies (Ab) can allow for direction of a cytotoxin to cancer cells; reducing side effects in healthy tissues (Table 1). Drugs can be linked to monoclonal antibodies (mAb) via cleavable peptides which release the active drug once it is taken up by cells into acidified compartments [12,13]. A limiting factor in developing these therapies can be discovery of cancer-specific antigens (Ag) to which the Ab-conjugated therapy may be targeted.
LIV-1 is an estrogen-regulated zinc transporter that is highly expressed in most breast cancers while its expression levels in other tissues is low [14,15]. Therefore, targeting chemotherapies to breast cancer cells has been achieved by conjugating the drug to humanized anti-LIV-1 Abs [15,16,17]. Another Ag that has been targeted in breast cancer is HER2 (human epidermal growth factor receptor-2), which when targeted, may concentrate therapies even in HER2-low cancers [15,18]. Many other Ab-drug conjugates are in the clinical stages of development including those specific for CD33 which is a sialic acid receptor that is over expressed in acute myeloid leukemia [19,20] and those specific for STEAP1 (six transmembrane epithelial antigen of the prostate 1) which is over expressed in prostate cancers [21]. Overall, the conjugation of cytotoxins to cancer-specific Abs has increased drug delivery to primary tumors and metastatic sites as well as cancers of the blood. Ab-drug conjugates have been approved by the FDA for treatment of Hodgkin lymphoma [22], HER2-positive breast cancer [23], acute myeloid leukaemia [24], and lymphoblastic leukaemia [25]; many others are currently in clinical trials for various diseases [26].
In addition to targeting receptors for location-specific concentration of therapies, controlled release of therapeutic substances also improves their efficacy and reduces off target effects. In some cases, non-internalizing targets on tumor cells are used to deliver Ab-drug conjugates to the tumor and any unbound complexes are cleared; next an activator is administered to release the drug from the antibody, allowing it to defuse into local tumor cells. This method reduces toxic side effects on healthy cells and increases efficacy of the drugs. It also allows for use of tumor targets that are not known to enhance uptake when engaged. This extracellular release of active drug can involve bioorthogonal, or “click-to-release”, reactions where a reaction between tetrazine and trans-cyclooctenes cause cleavage of allylic carbamates and release of the conjugated cytotoxin [27,28,29]. Work from the Robillard lab has demonstrated release of an antimitotic toxin from a diabody specific for tumor-associated glycoprotein-72 (gp72) when the tetrazine activator was administered to mice. Treatment with this technique resulted in increased anti-tumor activity in both ovarian and colon carcinoma models in mice; this was achieved without signs of off-target toxicity as the drug was selectively concentrated at the tumor site and was absent or present at low levels in other tissues [30]. Another form of “click to release” involves an isonitrile-based cleavage that was discovered by the Franzini lab and involves the rapid bioorthogonal removal of a 3-isocyanopropyl masking group through reaction with tetrazines [31,32,33]. Collectively, this method of selected drug release reduces side effects on healthy cells and increases potency of anti-cancer therapies.

2.2. Central Nervous System Therapies

Alzheimer’s disease is the most common form of dementia in the United States, where there are currently over 5 million people living with the disease [53]. Brain pathology characteristically involves the development of amyloid beta plaques, aggregated tau protein, neurofibrillary tangles, and a variety of neurological defects. As researchers have discovered more about Alzheimer’s disease, they have also unveiled promising targets for therapies [6]. However, a major limitation on delivery of neurodegenerative therapies is bioavailability and crossing the blood-brain-barrier (BBB). Fan et al. developed a brain-targeted nanoparticle (NP) that increases the delivery of curcumin, an anti-inflammatory compound [36]. Targeting of the curcumin-loaded NPs to the brain was achieved through conjugation of the particles to B6 peptides. B6 is a short peptide that binds the transferrin receptor (TfR) which is expressed on endothelial cells of the BBB and facilitates transcytosis; targeting to this receptor increases BBB-permeability and availability of B6-conjugates in the brain [36,54,55,56]. Administration of the B6-conjugated curcumin-loaded NP to mice improved their memory and spatial learning which has promise to extend to treatment of humans with neurodegenerative diseases [36].
Parkinson’s disease is another common neurodegenerative disorder that is characterized by the slowing of movements and the presence of a resting tremor [7,57]. As with Alzheimer’s disease, delivery of therapeutics for Parkinson’s disease also requires crossing the BBB. Rusiecka et al. enhanced the delivery of dopamine to the brain through conjugation to the cell-penetrating peptide TP10 [37] which is derived from wasp venom [58]. This heightened cell permeability improves delivery of TP10-conjugates to the brain and in the case of dopamine, delivers it in a form that has a high affinity for its receptor [37]. It should be noted that the lipophilic peptides such as TP10 are not inherently BBB specific and additional BBB-specific targeting modalities might enhance the specificity of this approach.
Additional examples of targeted CNS therapies include those focused on demyelination (a hallmark of multiple sclerosis-MS) and post-stroke, neuro-regeneration [38,39]. The aptamer LJM-3064 is a small (40 nt, ~13 kDa) single stranded DNA molecule selected in a screen for myelein-binding activity [59]. Tetramers of LJM-3064 administered to mice in a viral encephalitis model displayed significant remyelination of the spinal cord. The results indicate that the myelin-binding aptamer, when multimerized, has inherent pro-myelinogenic properties. LJM-3064 has subsequently been used to decorate the surfaces of anti-inflammatory exosomes (derived from mesenchymal stem cells). The aptamer-exosome bioconjugate reduced both CNS inflammation and demyelination in a murine experimental autoimmune encephalomyelitis (EAE) model of MS [38].
Neuro-regeneration by adult neural stem cells holds tremendous medical potential but has been limited, particularly in the context of ischemic stroke, by poor survival of migrating neuroblasts. In attempt to activate pro-survival pathways in these cells, the Akt-activating drug SC-79 has been encapsulated in NP decorated with Ab specific for a neural cell adhesion molecule (PSA-NECM). When the drug-loaded conjugates (α-PSA-NECM-NP-SC-79) were instilled into murine lateral ventricles, the authors noted activation of Akt in neuroblasts within in the subventricular zone [39]. Collectively, the ability to target conjugates for enhanced passage across the BBB and/or to specific sites of pathology shows promise for enhancing treatment of brain-specific diseases.

2.3. Antimicrobials

Since the discovery of penicillin by Alexander Fleming in the early 1900’s, antibiotics have been used to treat countless infections and have greatly improved medical care. However, emerging bacterial resistance to antibiotics is widely recognized and has hindered the effective use of many compounds [60,61]. The emergence of new and more extensively drug-resistant bacteria have encouraged the discovery of novel antibiotic compounds as well as targeted delivery of existing antibiotics which promotes more direct and concentrated administration of a drug [62]. By focusing delivery of the drug to the pathogen which is causing disease, the exposure of commensal microbes to antibiotic agents is reduced. This is important to prevent development of antibiotic resistance; in many cases the antibiotic activity of a drug is broad and sub-lethal exposure permits bacteria to enhance their drug resistance mechanisms. Therefore, targeting an antibiotic to a specific site or pathogen can both reduce generation of drug resistant microbes while leaving endogenous commensal (“good”) bacterial populations intact.
One method of targeting delivery of antibiotics and limiting off-target effects is to conjugate the antibiotic to a pathogen-specific Ab. Lehar et al. conjugated the antibiotic rifalogue to an Ab specific for MRSA (methicillin-resistant Staphylococcus aureus) and showed increased killing of MRSA in a murine bacteremia model. In this case, the antibiotic was not active until the conjugate-opsonized bacteria were engulfed in a phagosome, it was then activated by intracellular proteases and able to kill the bacteria within the host cell [40]. This method not only concentrates the antibiotic at the location of the pathogen, but also reduces exposure of commensal bacteria to the drug which would have the potential to induce antibiotic resistance mechanisms.
Similarly, targeted NPs and virus-like particles (VLPs) have also been used to concentrate antibiotic delivery. NPs enhance bioavailability, solubility, stability, and controlled release of different substances. In addition, due to their size, NPs are taken up by phagocytic mechanisms versus the passive entry of smaller molecules such as free drug [63,64]. Antibiotic-decorated particles have been shown to increase the amount of antibiotic internalized by lung-resident macrophages; therefore, concentrating the drug and potentially increasing effectiveness of treatments against stubborn pulmonary bacterial infections [65]. Gold NPs have also been utilized to enhance efficacy of antibiotics and reduce the necessary dose [66,67,68]. Work by Pornpattananangkul et al. utilized antibiotic-loaded gold NP that would release the drug upon contact with the pore-forming α-toxin of MRSA [41]. Since the particle did not liberate the antibiotic without the presence of toxin, the drug would only be released in sites near to the infection. Again, this concentrates the drug, increasing its activity and reducing off-target effects.

3. Targeted Vaccinations

Vaccinations have been used for centuries in order to protect populations against disease-causing microorganisms. In fact, the process of intentionally exposing an individual to a less virulent form of a pathogen has led to eradication of the disfiguring and deadly disease, small pox [69]. However, while this vaccine prevented later contraction of small pox, it initially caused mild disease as it involved administration of virulent cow pox virus [70]. Vaccine development has since evolved with an interest in increasing the safety of vaccinations without sacrificing protective efficacy. Inactivation of pathogens such as influenza [71] and cholera [72] greatly increase their safety for use as a vaccines. Safety is even further improved in vaccines against Hepatitis B [73], diptheria, and tetanus [74]; utilizing purified proteins or inactivated toxins in subunit vaccines: HBsAg and DTaP, respectively. Increasing the safety of these vaccines comes at a trade off with reduced efficacy since there is no live infection generating danger signals. In some cases, immune stimulation is restored through the addition of an adjuvant such as alum [75,76]. However, many adjuvants cause off-target effects, including unwanted activation of bystander cells and potential induction of autoimmunity [77,78]. These drawbacks to adjuvants have encouraged the development of adjuvant-independent enhancement of vaccines; this has been achieved through fusion of Ag to stimulatory or targeting molecules. Research aimed at generating a vaccine against a potential biological weapon, Francisella tularensis (Ft), has shown that opsonizing inactivated Ft with α-Ft Abs targets the bacteria to Fc receptors on immune cells. This interaction stimulates uptake and presentation of Ag, activation of immune cells, and ultimately results in production of an enhanced protective immune response against virulent Ft challenge [79,80,81]. Similar methods of targeting to host cells have been achieved through bioconjugation; we have diagrammed several of these in Figure 1.

3.1. Protein Subunit Vaccine Conjugates

Directing protein Ag to Ag-presenting cells (APCs) such as dendritic cells (DCs), increases both cellular and humoral immune responses. DCs phagocytose Ag and subsequently process and present peptides via both major histocompatibility complex-I (MHCI) and -II (MHCII), allowing for involvement of a variety of downstream cells. One example of targeting Ag to DCs is accomplished through the activating receptor, Clec9A (C-type lectin domain family 9 member A) [48,82,83]. Park et al. accomplished fusion of the influenza Ag M2e to α-Clec9A Ab through genetic engineering of a plasmid. Plasmids encoded the heavy and light chains of α-Clec9A Ab, a flexible glycine linker, and three repeats of the M2e peptide. The plasmids were then transfected into Freestyle 293F cells for expression of the Ag-Ab conjugates. The secreted Ag-Ab fusions were purified via protein G affinity chromatography. Ultimately, administration of the targeted Ags to mice resulted in increased specific immunoglobulin responses and increased protection against lethal influenza infection compared to Ag conjugated to an isotype control Ab [35]. Generation of vaccines via this method is diagramed in Figure 1a. Similar technologies have been used to fuse influenza hemagglutinin (HA) to α-MHC-II Ab, α-CCR1/3/5 Ab [42], or cytokines [43,44]. In the case of cytokines, when HA was conjugated to Xcl1 (a ligand for Xcr1) or Ccl3 (a ligand for CCR1/3/5) the vaccines targeted different subsets of DCs and therefore resulted in increased CD4+ and CD8+ T cell proliferation or only increased CD4+ T cell proliferation, respectively [43]. This phenomenon further emphasizes the ability to tailor targeting of Ags to induce specific immune responses through conjugation to different peptides. Further, these enhanced immune responses discussed above were seen in the absence of conventional adjuvant; when tested, targeted immunogens without adjuvant, were similarly or more efficient than untargeted-Ag administered with traditional adjuvant.
Conjugate vaccines have also been targeted to complement receptors on various immune cells. Pioneering work by Dempsey et al. involved conjugating hen egg lysozyme (HEL) to complement protein C3d, a ligand for complement receptors 2 (CR2) and 3 (CR3). Mice immunized with pure (C3d)3-HEL produced higher α-HEL Ab titers than those immunized with HEL plus Freud’s complete adjuvant [84]. Targeting Ag to CRs is a concept that our lab has also explored and applied to whole cell vaccines which will be discussed later in this review. CR2 is primarily expressed on B cells and follicular dendritic cells (FDCs); its engagement facilitates Ag uptake and presentation by FDCs, and activation of B cells. In addition, C3d engagement of CR3 on APCs induces translocation to the lymph node and increased Ag presentation. C3d conjugation has been applied to bacterial [49] as well as viral peptides [50,82] via genetic engineering of plasmids encoding the fusion constructs. This concept has also been extended to the minimum CR2-binding region of C3d, p28, which also has shown promise as a method for increasing Ag-specific immune responses [83,85,86,87,88].

3.2. Glyconjugate Vaccines

In addition to proteins, Ags of interest for vaccination may also be lipoproteins, peptidoglycan, polysaccharides (PS), and other capsular material. Enhancement of the immune response to PS components has been achieved by linking them to carrier or stimulatory peptides [89]. PS are primarily recognized by Toll-Like Receptors 2 (TLR2) and 4 (TLR4) which are expressed on many immune cells [90,91]. Linking PS to stimulatory or targeted peptides increases their recognition by the immune system; especially through interaction with helper T cells [45,92,93,94]. This technique has been applied to various vaccines through chemical conjugation (Figure 1d), including the approved vaccines against Neisseria meningitidis (Nm) and Streptococcus pneumoniae (Sp) [45].
To simplify and expand chemical conjugation, PS have been conjugated to carrier proteins or lipids through the use of glycosylation processes within bacteria [46,95,96,97]. In this case, engineered strains of Escherichia coli (Ec) that express oligosaccharyltransferases (OTase) are utilized for vaccine production. OTase transfers PS to carrier proteins and these conjugates can then be harvested and purified from Ec (Figure 1b). An example of this conjugation technique involves Ags of Klebsiella pneumoniae (Kp), a multi-drug-resistant bacteria that produces substantial amounts of PS capsule which shields it from the immune system [98]. Development of a PS capsule-targeting immune response, as opposed to a protein-targeted response would be extremely advantageous for this pathogen. A bivalent vaccine against two Kp serotypes has been engineered by Feldman et al. using both K1 and K2 capsular PS locus clusters [46]. These PS were each produced by Ec which transferred them to exotoxin protein A from Pseudomonas aeruginosa (Pa) as a carrier. These purified K1 and K2 conjugates were then mixed and used to vaccinate mice; the bivalent vaccine induced increased α-Kp Ab levels as well as protection from lethal Kp infection.

3.3. Conjugated Nanoparticle-Based Vaccines

Nanoparticles are larger than subunit vaccines and have been shown to promote trafficking of Ag to lymph nodes [99]. NPs can be formed using a variety of materials including: self-assembling lipids (liposomes), sucrose polymers, gold, and proteins [100]. Peptide NPs may be loaded by using genetic techniques, leaving their self-assembling domains intact [101]. In addition, NPs may be decorated via chemical conjugation (Figure 1c). This process involves activation of the NP to reveal active residues that are then available for bond formation with moieties of interest [102]. For example, N-hydroxy-succinimide is used to activate primary amines in lysine residues [102,103]. This strategy was utilized to display Plasmodium falciparum protein Pfs25 on NPs to be tested as a malaria vaccine [47,104]. It was found that chemical conjugation of the Ag to the NP resulted in generation of more Abs, compared to genetic or SpyTag conjugation where “tag” and “catcher” peptides are added to the desired conjugates. In some cases, the conjugated cargo may be released from the NPs in a regulated manner. Schudel et al. recently utilized NPs to concentrate delivery at the lymph nodes where cargo was then released in phases due to the use of linkers with different half-lives [105]. Delaying the release of some cargo from NPs may allow for engagement of different lymphatic cell populations and has the potential to enhance immune responses and/or drug effects. In addition to targeting the lymph nodes, loaded NPs have also been further decorated with mannose [48] and anti-DEC-205 mAbs [106]; targeting them to mannose receptor on APCs or DEC-205 on DCs, respectively.

3.4. Targeting and Conjugation Involving Whole-Cell Vaccines

Pompa-Mera et al. created a plasmid which encoded a fusion protein of p28, a Trichinella spiralis (Ts) peptide, and a Salmonella outer membrane protein (OMP) with a proteolytic site between the OMP and p28-Ts peptide fusion [52]. The plasmid was then transformed into Salmonella, resulting in a strain that produced the OM-located fusion protein, from which the targeted Ts peptide was liberated. This strain was administered as a live vaccine to mice where it induced increased Ts-specific Ab levels and conferred protection against subsequent Ts challenge. This method of vaccine generation is diagramed in Figure 1e and combines the targeting ability of p28 with the generation of danger signals from a live infection.
Our lab similarly produced a CR-targeted vaccine via generation of a plasmid encoding a CR-targeting fusion protein that was transformed into bacteria which were then administered as a whole cell vaccine. In this case, we engineered a plasmid encoding C3d conjugated to YadA, a well-characterized Yersinia OMP without a cleavage site between the proteins. Therefore, transformation of bacteria with the plasmid resulted in expression of OM-localized, surface-displayed C3d. This promotes targeting of the entire bacterial immunogen to CRs on immune cells and enhances protective responses; we have demonstrated this utilizing murine pulmonary Francisella tularensis infection as a model [51]. This technique is diagramed in Figure 1f.

4. Summary

The ability to combine therapies or prophylactics with another element that facilitates delivery to their target has advanced a wide range of fields; including the treatment of cancers, dementia, and bacterial infections, as well as enhancement of vaccinations. Targeting has been achieved through bioconjugation of two or more moieties that embody different functions. Generally, one subunit will provide the targeting or enhancement function, whereas the other performs the desired treatment. This not only enhances treatment efficacy but also limits unwanted side effects; appealing traits to virtually all patient care. It is important to note however, that the potential for side effects may not be absolutely eliminated, even with targeting to distinct sites or cells. Another potential drawback in developing targeted treatments is the delay in approval for new treatments. In the case of cancer treatments, a broadly administered chemotherapy agent may be more readily available for patient care. Further, targeting therapies and prophylactic treatments to specific cell types may impede involvement of other cells whose benefits are not yet fully realized. These facts underscore the need for continued research and advancement of targeted therapies.

Author Contributions

K.M.T. and K.R.O.H. wrote and edited manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This review was funded by grants from the National Institutes of Health (RO1AI123129) and the Department of Defense (W81XH1910661).

Acknowledgments

We wish to thank Lauren Shoudy and Sudeep Kumar for their review of this work.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the writing of the manuscript.

References

  1. Muhamad, N.; Plengsuriyakarn, T.; Na-Bangchang, K. Application of active targeting nanoparticle delivery system for chemotherapeutic drugs and traditional/herbal medicines in cancer therapy: A systematic review. Int. J. Nanomed. 2018, 13, 3921–3935. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Cohn, L.; Delamarre, L. Dendritic cell-targeted vaccines. Front. Immunol. 2014, 5, 255. [Google Scholar] [CrossRef] [PubMed]
  3. Goyvaerts, C.; Breckpot, K. Pros and Cons of Antigen-Presenting Cell Targeted Tumor Vaccines. J. Immunol Res. 2015, 2015, 785634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Liu, X.; Liu, S.; Lyu, H.; Riker, A.I.; Zhang, Y.; Liu, B. Development of Effective Therapeutics Targeting HER3 for Cancer Treatment. Biol. Proced. Online 2019, 21, 5. [Google Scholar] [CrossRef] [Green Version]
  5. Sancho, D.; Mourao-Sa, D.; Joffre, O.P.; Schulz, O.; Rogers, N.C.; Pennington, D.J.; Carlyle, J.R.; Reis e Sousa, C. Tumor therapy in mice via antigen targeting to a novel, DC-restricted C-type lectin. J. Clin. Investig. 2008, 118, 2098–2110. [Google Scholar] [CrossRef] [PubMed]
  6. Loera-Valencia, R.; Cedazo-Minguez, A.; Kenigsberg, P.; Page, G.; Duarte, A.; Giusti, P.; Zusso, M.; Robert, P.; Frisoni, G.; Cattaneo, A.; et al. Current and emerging avenues for Alzheimer’s disease drug targets. J. Intern. Med. 2019, 286, 398–437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Beitz, J. Parkinson’s disease: A review. Front. Biosci. (Sch. Ed.) 2014, 6, 65–74. [Google Scholar] [CrossRef]
  8. Nguyen, L.; Garcia, J.; Gruenberg, K.; MacDougall, C. Multidrug-Resistant Pseudomonas Infections: Hard to Treat, But Hope on the Horizon? Curr. Infect. Dis. Rep. 2018, 20, 23. [Google Scholar] [CrossRef]
  9. Magiorakos, A.P.; Srinivasan, A.; Carey, R.B.; Carmeli, Y.; Falagas, M.E.; Giske, C.G.; Harbarth, S.; Hindler, J.F.; Kahlmeter, G.; Olsson-Liljequist, B.; et al. Multidrug-resistant, Extensively Drug-Resistant and Pandrug-Resistant Bacteria: An International Expert Proposal for Interim Standard Definitions for Acquired Resistance. Clin. Microbiol. Infect. Off. Publ. Eur. Soc. Clin. Microbiol. Infect. Dis. 2012, 18, 268–281. [Google Scholar] [CrossRef] [Green Version]
  10. DeVita, V.; Chu, E. A history of cancer chemotherapy. Cancer Res. 2008, 68, 8643–8653. [Google Scholar] [CrossRef] [Green Version]
  11. Cheung-Ong, K.; Giaever, G.; Nislow, C. DNA-damaging agents in cancer chemotherapy: Serendipity and chemical biology. Chem. Biol. 2013, 20, 648–659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Chari, R.; Miller, M.; Widdison, W. Antibody-drug conjugates: An emerging concept in cancer therapy. Angew. Chem. (Int. Ed. Engl.) 2014, 53, 3796–3827. [Google Scholar] [CrossRef] [PubMed]
  13. Panowski, S.; Bhakta, S.; Raab, H.; Polakis, P.; Junutula, J. Site-specific antibody drug conjugates for cancer therapy. In mAbs; Taylor Francis: Abingdon-on-Thames, UK, 2014; Volume 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Grattan, B.; Freake, H. Zinc and cancer: Implications for LIV-1 in breast cancer. Nutrients 2012, 4, 648–675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Lyons, T. Targeted Therapies for Triple-Negative Breast Cancer. Curr. Treat. Options Oncol. 2019, 20. [Google Scholar] [CrossRef] [PubMed]
  16. Modi, S.; Pusztai, L.; Forero, A.; Mita, M.; Miller, K.; Weise, A.; Krop, I.; Burris, H.; Kalinsky, K.; Tsai, M.; et al. Abstract PD3-14: Phase 1 study of the antibody-drug conjugate SGN-LIV1A in patients with heavily pretreated triple-negative metastatic breast cancer. Cancer Res. 2018. [Google Scholar] [CrossRef]
  17. McGuinness, J.; Kalinsky, K. Antibody-drug conjugates in metastatic triple negative breast cancer: A spotlight on sacituzumab govitecan, ladiratuzumab vedotin, and trastuzumab deruxtecan. Expert Opin. Biol. Ther. 2020. [Google Scholar] [CrossRef] [PubMed]
  18. Xu, Z.; Guo, D.; Jiang, Z.; Tong, R.; Jiang, P.; Bai, L.; Chen, L.; Zhu, Y.; Guo, C.; Shi, J.; et al. Novel HER2-Targeting Antibody-Drug Conjugates of Trastuzumab Beyond T-DM1 in Breast Cancer: Trastuzumab Deruxtecan(DS-8201a) and (Vic-)Trastuzumab Duocarmazine (SYD985). Eur. J. Med. Chem. 2019, 183, 111682. [Google Scholar] [CrossRef] [PubMed]
  19. De Propris, M.; Raponi, S.; Diverio, D.; Milani, M.; Meloni, G.; Falini, B.; Foà, R.; Guarini, A. High CD33 expression levels in acute myeloid leukemia cells carrying the nucleophosmin (NPM1) mutation. Haematologica 2011, 96, 1548. [Google Scholar] [CrossRef] [Green Version]
  20. Hughes, B. Antibody-drug conjugates for cancer: Poised to deliver? Nat. Rev. Drug Discov. 2010, 9, 665. [Google Scholar] [CrossRef]
  21. Gomes, I.; Arinto, P.; Lopes, C.; Santos, C.; Maia, C. STEAP1 is overexpressed in prostate cancer and prostatic intraepithelial neoplasia lesions, and it is positively associated with Gleason score. In Urologic Oncology: Seminars and Original Investigations; Elsevier: Hoboken, NJ, USA, 2014; Volume 32. [Google Scholar] [CrossRef] [PubMed]
  22. Richardson, N.; Kasamon, Y.; Chen, H.; de Claro, R.; Ye, J.; Blumenthal, G.; Farrell, A.; Pazdur, R. FDA Approval Summary: Brentuximab Vedotin in First-Line Treatment of Peripheral T-Cell Lymphoma. Oncology 2019, 24, e180. [Google Scholar] [CrossRef] [Green Version]
  23. Wedam, S.; Fashoyin-Aje, L.; Gao, X.; Bloomquist, E.; Tang, S.; Sridhara, R.; Goldberg, K.; King-Kallimanis, B.; Theoret, M.; Ibrahim, A.; et al. FDA Approval Summary: Ado-Trastuzumab Emtansine for the Adjuvant Treatment of HER2-positive Early Breast Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2020, 26, 4180–4185. [Google Scholar] [CrossRef] [Green Version]
  24. Norsworthy, K.; Ko, C.; Lee, J.; Liu, J.; John, C.; Przepiorka, D.; Farrell, A.; Pazdur, R. FDA Approval Summary: Mylotarg for Treatment of Patients with Relapsed or Refractory CD33-Positive Acute Myeloid Leukemia. Oncology 2018, 23, 1103. [Google Scholar] [CrossRef] [Green Version]
  25. Lamb, Y. Inotuzumab Ozogamicin: First Global Approval. Drugs 2017, 77, 1603–1610. [Google Scholar] [CrossRef]
  26. Lambert, J.; Morris, C. Antibody-Drug Conjugates (ADCs) for Personalized Treatment of Solid Tumors: A Review. Adv. Ther. 2017, 34, 1015–1035. [Google Scholar] [CrossRef] [Green Version]
  27. Versteegen, R.; Rossin, R.; ten Hoeve, W.; Janssen, H.; Robillard, M. Click to release: Instantaneous doxorubicin elimination upon tetrazine ligation. Angew. Chem. (Int. Ed. Engl.) 2013, 52, 14362–14366. [Google Scholar] [CrossRef]
  28. Oliveira, B.; Guo, Z.; Bernardes, G. Inverse electron demand Diels-Alder reactions in chemical biology. Chem. Soc. Rev. 2017, 46, 4895–4950. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Van Onzen, A.; Versteegen, R.; Hoeben, F.; Filot, I.; Rossin, R.; Zhu, T.; Wu, J.; Hudson, P.; Janssen, H.; Ten Hoeve, W.; et al. Bioorthogonal Tetrazine Carbamate Cleavage by Highly Reactive trans-Cyclooctene. J. Am. Chem. Soc. 2020, 142, 10955–10963. [Google Scholar] [CrossRef]
  30. Rossin, R.; Versteegen, R.; Wu, J.; Khasanov, A.; Wessels, H.; Steenbergen, E.; Ten Hoeve, W.; Janssen, H.; van Onzen, A.; Hudson, P.; et al. Chemically triggered drug release from an antibody-drug conjugate leads to potent antitumour activity in mice. Nat. Commun. 2018, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Tu, J.; Xu, M.; Parvez, S.; Peterson, R.; Franzini, R. Bioorthogonal Removal of 3-Isocyanopropyl Groups Enables the Controlled Release of Fluorophores and Drugs in Vivo. J. Am. Chem. Soc. 2018, 140, 8410–8414. [Google Scholar] [CrossRef] [PubMed]
  32. Xu, M.; Deb, T.; Tu, J.; Franzini, R. Tuning Isonitrile/Tetrazine Chemistry for Accelerated Deprotection and Formation of Stable Conjugates. J. Org. Chem. 2019, 84, 15520–15529. [Google Scholar] [CrossRef]
  33. Tu, J.; Svatunek, D.; Parvez, S.; Eckvahl, H.; Xu, M.; Peterson, R.; Houk, K.; Franzini, R. Isonitrile-responsive and bioorthogonally removable tetrazine protecting groups. Chem. Sci. 2019, 11, 169–179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Sussman, D.; Smith, L.; Anderson, M.; Duniho, S.; Hunter, J.; Kostner, H.; Miyamoto, J.; Nesterova, A.; Westendorf, L.; Van Epps, H.; et al. SGN-LIV1A: A novel antibody-drug conjugate targeting LIV-1 for the treatment of metastatic breast cancer. Mol. Cancer Ther. 2014, 13, 2991–3000. [Google Scholar] [CrossRef] [Green Version]
  35. Park, H.Y.; Tan, P.S.; Kavishna, R.; Ker, A.; Lu, J.; Chan, C.E.Z.; Hanson, B.J.; MacAry, P.A.; Caminschi, I.; Shortman, K.; et al. Enhancing vaccine antibody responses by targeting Clec9A on dendritic cells. NPJ Vaccines 2017, 2, 31. [Google Scholar] [CrossRef] [Green Version]
  36. Fan, S.; Zheng, Y.; Liu, X.; Fang, W.; Chen, X.; Liao, W.; Jing, X.; Lei, M.; Tao, E.; Ma, Q.; et al. Curcumin-loaded PLGA-PEG nanoparticles conjugated with B6 peptide for potential use in Alzheimer’s disease. Drug Deliv. 2018, 25, 1091–1102. [Google Scholar] [CrossRef] [Green Version]
  37. Rusiecka, I.; Ruczyński, J.; Kozłowska, A.; Backtrog, E.; Mucha, P.; Kocić, I.; Rekowski, P. TP10-Dopamine Conjugate as a Potential Therapeutic Agent in the Treatment of Parkinson’s Disease. Bioconjugate Chem. 2019, 30, 760–774. [Google Scholar] [CrossRef] [PubMed]
  38. Hosseini Shamili, F.; Alibolandi, M.; Rafatpanah, H.; Abnous, K.; Mahmoudi, M.; Kalantari, M.; Taghdisi, S.; Ramezani, M. Immunomodulatory properties of MSC-derived exosomes armed with high affinity aptamer toward mylein as a platform for reducing multiple sclerosis clinical score. J. Control. Release Off. J. Control. Release Soc. 2019, 299, 149–164. [Google Scholar] [CrossRef] [PubMed]
  39. Balasubramanian, V.; Domanskyi, A.; Renko, J.; Sarparanta, M.; Wang, C.; Correia, A.; Mäkilä, E.; Alanen, O.; Salonen, J.; Airaksinen, A.; et al. Engineered antibody-functionalized porous silicon nanoparticles for therapeutic targeting of pro-survival pathway in endogenous neuroblasts after stroke. Biomaterials 2020, 227, 119556. [Google Scholar] [CrossRef]
  40. Lehar, S.; Pillow, T.; Xu, M.; Staben, L.; Kajihara, K.; Vandlen, R.; DePalatis, L.; Raab, H.; Hazenbos, W.; Morisaki, J.; et al. Novel antibody-antibiotic conjugate eliminates intracellular S. aureus. Nature 2015, 527, 323–328. [Google Scholar] [CrossRef] [PubMed]
  41. Pornpattananangkul, D.; Zhang, L.; Olson, S.; Aryal, S.; Obonyo, M.; Vecchio, K.; Huang, C.; Zhang, L. Bacterial toxin-triggered drug release from gold nanoparticle-stabilized liposomes for the treatment of bacterial infection. J. Am. Chem. Soc. 2011, 133, 4132–4139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Grødeland, G.; Mjaaland, S.; Tunheim, G.; Fredriksen, A.; Bogen, B. The specificity of targeted vaccines for APC surface molecules influences the immune response phenotype. PLoS ONE 2013, 8, e0080008. [Google Scholar] [CrossRef] [Green Version]
  43. Lysen, A.; Braathen, R.; Gudjonsson, A.; Tesfaye, D.Y.; Bogen, B.; Fossum, E. Dendritic cell targeted Ccl3- and Xcl1-fusion DNA vaccines differ in induced immune responses and optimal delivery site. Sci. Rep. 2019, 9, 1820. [Google Scholar] [CrossRef] [PubMed]
  44. Fossum, E.; Grødeland, G.; Terhorst, D.; Tveita, A.; Vikse, E.; Mjaaland, S.; Henri, S.; Malissen, B.; Bogen, B. Vaccine molecules targeting Xcr1 on cross-presenting DCs induce protective CD8+ T-cell responses against influenza virus. Eur. J. Immunol. 2015, 45, 624–635. [Google Scholar] [CrossRef] [PubMed]
  45. Berti, F.; Micoli, F. Improving efficacy of glycoconjugate vaccines: From chemical conjugates to next generation constructs. Curr. Opin. Immunol. 2020, 65, 42–49. [Google Scholar] [CrossRef] [PubMed]
  46. Feldman, M.F.; Mayer Bridwell, A.E.; Scott, N.E.; Vinogradov, E.; McKee, S.R.; Chavez, S.M.; Twentyman, J.; Stallings, C.L.; Rosen, D.A.; Harding, C.M. A promising bioconjugate vaccine against hypervirulent Klebsiella pneumoniae. Proc. Natl. Acad. Sci. USA 2019, 116, 18655–18663. [Google Scholar] [CrossRef] [Green Version]
  47. Leneghan, D.B.; Miura, K.; Taylor, I.J.; Li, Y.; Jin, J.; Brune, K.D.; Bachmann, M.F.; Howarth, M.; Long, C.A.; Biswas, S. Nanoassembly routes stimulate conflicting antibody quantity and quality for transmission-blocking malaria vaccines. Nature 2017. [Google Scholar] [CrossRef]
  48. Layek, B.; Lipp, L.; Singh, J. APC targeted micelle for enhanced intradermal delivery of hepatitis B DNA vaccine. J. Control. Release 2015, 207, 143–153. [Google Scholar] [CrossRef]
  49. Musa, H.H.; Zhang, W.J.; Lv, J.; Duan, X.L.; Yang, Y.; Zhu, C.H.; Li, H.F.; Chen, K.W.; Meng, X.; Zhu, G.Q. The molecular adjuvant mC3d enhances the immunogenicity of FimA from type I fimbriae of Salmonella enterica serovar Enteritidis. J. Microbiol. Immunol. Infect. 2014, 47, 57–62. [Google Scholar] [CrossRef] [Green Version]
  50. Green, T.D.; Montefiori, D.C.; Ross, T.M. Enhancement of antibodies to the human immunodeficiency virus type 1 envelope by using the molecular adjuvant C3d. J. Virol. 2003, 77, 2046–2055. [Google Scholar] [CrossRef] [Green Version]
  51. Holland-Tummillo, K.M.; Shoudy, L.E.; Steiner, D.; Kumar, S.; Rosa, S.J.; Namjoshi, P.; Singh, A.; Sellati, T.J.; Gosselin, E.J.; Hazlett, K.R. Autotransporter-Mediated Display of Complement Receptor Ligands by Gram-Negative Bacteria Increases Antibody Responses and Limits Disease Severity. Pathogens 2020, 9, 375. [Google Scholar] [CrossRef] [PubMed]
  52. Pompa-Mera, E.N.; Arroyo-Matus, P.; Ocana-Mondragon, A.; Gonzalez-Bonilla, C.R.; Yepez-Mulia, L. Protective immunity against enteral stages of Trichinella spiralis elicited in mice by live attenuated Salmonella vaccine that secretes a 30-mer parasite epitope fused to the molecular adjuvant C3d-P28. Res. Vet. Sci. 2014, 97, 533–545. [Google Scholar] [CrossRef] [PubMed]
  53. 2016 Alzheimer’s disease facts and figures. Alzheimer’s Dement. J. Alzheimer’s Assoc. 2016, 12. [CrossRef]
  54. Xia, H.; Anderson, B.; Mao, Q.; Davidson, B. Recombinant human adenovirus: Targeting to the human transferrin receptor improves gene transfer to brain microcapillary endothelium. J. Virol. 2000, 74, 11359–11366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Prades, R.; Guerrero, S.; Araya, E.; Molina, C.; Salas, E.; Zurita, E.; Selva, J.; Egea, G.; López-Iglesias, C.; Teixidó, M.; et al. Delivery of gold nanoparticles to the brain by conjugation with a peptide that recognizes the transferrin receptor. Biomaterials 2012, 33, 7194–7205. [Google Scholar] [CrossRef]
  56. Johnsen, K.; Burkhart, A.; Thomsen, L.; Andresen, T.; Moos, T. Targeting the transferrin receptor for brain drug delivery. Prog. Neurobiol. 2019, 181, 101665. [Google Scholar] [CrossRef] [PubMed]
  57. Reich, S.; Savitt, J. Parkinson’s Disease. Med. Clin. N. Am. 2019, 103, 337–350. [Google Scholar] [CrossRef]
  58. Yandek, L.; Pokorny, A.; Florén, A.; Knoelke, K.; Langel, U.; Almeida, P. Mechanism of the cell-penetrating peptide transportan 10 permeation of lipid bilayers. Biophys. J. 2007, 92, 2434–2444. [Google Scholar] [CrossRef] [Green Version]
  59. Nastasijevic, B.; Wright, B.; Smestad, J.; Warrington, A.; Rodriguez, M.; Maher, L. Remyelination induced by a DNA aptamer in a mouse model of multiple sclerosis. PLoS ONE 2012, 7, e0039595. [Google Scholar] [CrossRef] [Green Version]
  60. Khardori, N.; Stevaux, C.; Ripley, K. Antibiotics: From the Beginning to the Future: Part 1. Indian J. Pediatrics 2020, 87, 39–42. [Google Scholar] [CrossRef]
  61. Mohr, K. History of Antibiotics Research. Curr. Top. Microbiol. Immunol. 2016, 398, 237–272. [Google Scholar] [CrossRef]
  62. Davies, J.; Davies, D. Origins and evolution of antibiotic resistance. Microbiol. Mol. Biol. Rev. 2010, 74, 417–433. [Google Scholar] [CrossRef] [Green Version]
  63. Mudshinge, S.; Deore, A.; Patil, S.; Bhalgat, C. Nanoparticles: Emerging carriers for drug delivery. Saudi Pharm. J. SPJ Off. Publ. Saudi Pharm. Soc. 2011, 19, 129–141. [Google Scholar] [CrossRef] [Green Version]
  64. Gelperina, S.; Kisich, K.; Iseman, M.; Heifets, L. The potential advantages of nanoparticle drug delivery systems in chemotherapy of tuberculosis. Am. J. Respir. Crit. Care Med. 2005, 172, 1487–4392. [Google Scholar] [CrossRef] [Green Version]
  65. Crooke, S.; Schimer, J.; Raji, I.; Wu, B.; Oyelere, A.; Finn, M. Lung Tissue Delivery of Virus-Like Particles Mediated by Macrolide Antibiotics. Mol. Pharm. 2019, 16, 2947–2955. [Google Scholar] [CrossRef] [PubMed]
  66. Yang, P.; Pageni, P.; Rahman, M.; Bam, M.; Zhu, T.; Chen, Y.; Nagarkatti, M.; Decho, A.; Tang, C. Gold Nanoparticles with Antibiotic-Metallopolymers toward Broad-Spectrum Antibacterial Effects. Adv. Healthc. Mater. 2019, 8, 1800854. [Google Scholar] [CrossRef] [PubMed]
  67. Bagga, P.; Siddiqui, H.; Akhtar, J.; Mahmood, T.; Zahera, M.; Khan, M. Gold Nanoparticles Conjugated Levofloxacin: For Improved Antibacterial Activity over Levofloxacin Alone. Curr. Drug Deliv. 2017, 14, 1114–1119. [Google Scholar] [CrossRef]
  68. Palmieri, G.; Tatè, R.; Gogliettino, M.; Balestrieri, M.; Rea, I.; Terracciano, M.; Proroga, Y.; Capuano, F.; Anastasio, A.; De Stefano, L. Small Synthetic Peptides Bioconjugated to Hybrid Gold Nanoparticles Destroy Potentially Deadly Bacteria at Submicromolar Concentrations. Bioconjugate Chem. 2018, 29, 3877–3885. [Google Scholar] [CrossRef] [PubMed]
  69. Zhang, Y.; Zhao, K. Global Eradication of Small-Pox: Historical Fact, Experiences and Enlightenment. Zhonghua Liu Xing Bing Xue Za Zhi 1999, 20, 67–70. [Google Scholar]
  70. Baxby, D. Edward Jenner’s Inquiry; A Bicentenary Analysis. Vaccine 1999, 17, 301–307. [Google Scholar] [CrossRef]
  71. Buckland, B.C. The development and manufacture of influenza vaccines. Hum. Vaccin Immunother. 2015, 11, 1357–1360. [Google Scholar] [CrossRef] [PubMed]
  72. Harris, J.B. Cholera: Immunity and Prospects in Vaccine Development. J. Infect. Dis. 2018, 218, S141–S146. [Google Scholar] [CrossRef] [Green Version]
  73. Gerlich, W.H. Medical Virology of Hepatitis B: How it began and where we are now. Virol. J. 2013, 10, 239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Ogden, S.A.; Ludlow, J.T.; Alsayouri, K. Diphtheria Tetanus Pertussis (DTaP) Vaccine. 2020. Available online: https://www.ncbi.nlm.nih.gov/books/NBK545173/ (accessed on 18 December 2020).
  75. Harandi, A.M. Systems Analysis of Human Vaccine Adjuvants. Semin. Immunol. 2018, 39, 30–34. [Google Scholar] [CrossRef]
  76. Del Giudice, G.; Rappuoli, R.; Didierlaurent, A.M. Correlates of adjuvanticity: A review on adjuvants in licensed vaccines. Semin. Immunol. 2018, 39, 14–21. [Google Scholar] [CrossRef]
  77. Di Pasquale, A.; Preiss, S.; Tavares Da Silva, F.; Garcon, N. Vaccine Adjuvants: From 1920 to 2015 and Beyond. Vaccines 2015, 3, 320–343. [Google Scholar] [CrossRef] [Green Version]
  78. Kool, M.; Fierens, K.; Lambrecht, B. Alum Adjuvant: Some of the Tricks of the Oldest Adjuvant. J. Med Microbiol. 2012, 61. [Google Scholar] [CrossRef] [Green Version]
  79. Rawool, D.B.; Bitsaktsis, C.; Li, Y.; Gosselin, D.R.; Lin, Y.; Kurkure, N.V.; Metzger, D.W.; Gosselin, E.J. Utilization of Fc receptors as a mucosal vaccine strategy against an intracellular bacterium, Francisella tularensis. J. Immunol. 2008, 180, 5548–5557. [Google Scholar] [CrossRef]
  80. Gosselin, E.J.; Bitsaktsis, C.; Li, Y.; Iglesias, B.V. Fc receptor-targeted mucosal vaccination as a novel strategy for the generation of enhanced immunity against mucosal and non-mucosal pathogens. Arch. Immunol. Ther. Exp. 2009, 57, 311–323. [Google Scholar] [CrossRef]
  81. Iglesias, B.; Bitsaktsis, C.; Pham, G.; Drake, J.; Hazlett, K.; Porter, K.; Gosselin, E. Multiple mechanisms mediate enhanced immunity generated by mAb-inactivated F. tularensis immunogen. Immunol. Cell Biol. 2013, 91, 139–148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Haas, K.M.; Toapanta, F.R.; Oliver, J.A.; Poe, J.C.; Weis, J.H.; Karp, D.R.; Bower, J.F.; Ross, T.M.; Tedder, T.F. Cutting edge: C3d functions as a molecular adjuvant in the absence of CD21/35 expression. J. Immunol 2004, 172, 5833–5837. [Google Scholar] [CrossRef] [Green Version]
  83. Bower, J.F.; Ross, T.M. A minimum CR2 binding domain of C3d enhances immunity following vaccination. Adv. Exp. Med. Biol. 2006, 586, 249–264. [Google Scholar]
  84. Dempsey, P.W.; Allison, M.E.; Akkaraju, S.; Goodnow, C.C.; Fearon, D.T. C3d of complement as a molecular adjuvant: Bridging innate and acquired immunity. Science 1996, 271, 348–350. [Google Scholar] [CrossRef] [Green Version]
  85. García-Machorro, J.; López-González, M.; Barrios-Rojas, O.; Fernández-Pomares, C.; Sandoval-Montes, C.; Santos-Argumedo, L.; Villegas-Sepúlveda, N.; Gutiérrez-Castañeda, B.; Cedillo-Barrón, L. DENV-2 Subunit Proteins Fused to CR2 Receptor-Binding Domain (P28)-induces Specific and Neutralizing Antibodies to the Dengue Virus in Mice. Hum. Vaccines Immunother. 2013, 9, 2326–2335. [Google Scholar] [CrossRef] [Green Version]
  86. Dunn, M.D.; Rossi, S.L.; Carter, D.M.; Vogt, M.R.; Mehlhop, E.; Diamond, M.S.; Ross, T.M. Enhancement of anti-DIII Antibodies by the C3d Derivative P28 Results in Lower Viral Titers and Augments Protection in Mice. Virol. J. 2010, 7, 95. [Google Scholar] [CrossRef] [Green Version]
  87. Wang, L.X.; Xu, W.; Guan, Q.D.; Chu, Y.W.; Wang, Y.; Xiong, S.D. Contribution of C3d-P28 Repeats to Enhancement of Immune Responses Against HBV-preS2/S Induced by Gene Immunization. World J. Gastroenterol. 2004, 10, 2072–2077. [Google Scholar] [CrossRef] [PubMed]
  88. Zhang, D.; Xia, Q.; Wu, J.; Liu, D.; Wang, X.; Niu, Z. Construction and Immunogenicity of DNA Vaccines Encoding Fusion Protein of Murine Complement C3d-p28 and GP5 Gene of Porcine Reproductive and Respiratory Syndrome Virus. Vaccine 2011, 29, 629–635. [Google Scholar] [CrossRef] [PubMed]
  89. Mettu, R.; Chen, C.; Wu, C. Synthetic carbohydrate-based vaccines: Challenges and opportunities. J. Biomed. Sci. 2020, 27. [Google Scholar] [CrossRef] [Green Version]
  90. Barton, G.; Medzhitov, R. Toll-like receptor signaling pathways. Science 2003, 300, 1524–1525. [Google Scholar] [CrossRef] [PubMed]
  91. Wang, Q.; McLoughlin, R.; Cobb, B.; Charrel-Dennis, M.; Zaleski, K.; Golenbock, D.; Tzianabos, A.; Kasper, D. A bacterial carbohydrate links innate and adaptive responses through Toll-like receptor 2. J. Exp. Med. 2006, 203, 2853–2863. [Google Scholar] [CrossRef] [Green Version]
  92. Test, S.T.; Mitsuyoshi, J.; Connolly, C.C.; Lucas, A.H. Increased immunogenicity and induction of class switching by conjugation of complement C3d to pneumococcal serotype 14 capsular polysaccharide. Infect. Immun. 2001, 69, 3031–3040. [Google Scholar] [CrossRef] [Green Version]
  93. Riddle, M.; Kaminski, R.; Di Paolo, C.; Porter, C.; Gutierrez, R.; Clarkson, K.; Weerts, H.; Duplessis, C.; Castellano, A.; Alaimo, C.; et al. Safety and Immunogenicity of a Candidate Bioconjugate Vaccine against Shigella flexneri 2a Administered to Healthy Adults: A Single-Blind, Randomized Phase I Study. Clin. Vaccine Immunol. Civ. 2016, 23, 908–917. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Rappuoli, R. Glycoconjugate vaccines: Principles and mechanisms. Sci. Transl. Med. 2018, 10. [Google Scholar] [CrossRef]
  95. Wacker, M.; Linton, D.; Hitchen, P.; Nita-Lazar, M.; Haslam, S.; North, S.; Panico, M.; Morris, H.; Dell, A.; Wren, B.; et al. N-linked glycosylation in Campylobacter jejuni and its functional transfer into E. coli. Science 2002, 298, 1790–1793. [Google Scholar] [CrossRef] [PubMed]
  96. Ravenscroft, N.; Haeuptle, M.; Kowarik, M.; Fernandez, F.; Carranza, P.; Brunner, A.; Steffen, M.; Wetter, M.; Keller, S.; Ruch, C.; et al. Purification and characterization of a Shigella conjugate vaccine, produced by glycoengineering Escherichia coli. Glycobiology 2016, 26, 51–62. [Google Scholar] [CrossRef] [Green Version]
  97. Feldman, M.; Wacker, M.; Hernandez, M.; Hitchen, P.; Marolda, C.; Kowarik, M.; Morris, H.; Dell, A.; Valvano, M.; Aebi, M. Engineering N-linked protein glycosylation with diverse O antigen lipopolysaccharide structures in Escherichia coli. Proc. Natl. Acad. Sci. USA 2005, 102, 3016–3021. [Google Scholar] [CrossRef] [Green Version]
  98. Doorduijn, D.J.; Rooijakkers, S.H.; van Schaik, W.; Bardoel, B.W. Complement Resistance Mechanisms of Klebsiella Pneumoniae. Immunobiology 2016, 221, 1102–1109. [Google Scholar] [CrossRef] [Green Version]
  99. Tokatlian, T.; Read, B.J.; Jones, C.A.; Kulp, D.W.; Menis, S.; Chang, J.Y.H.; Steichen, J.M.; Kumari, S.; Allen, J.; Dane, E.L.; et al. Innate Immune Recognition of Glycans Targets HIV Nanoparticle Immunogens to Germinal Centers. Science 2019, 363, 649–654. [Google Scholar] [CrossRef] [PubMed]
  100. Wang, E.C.; Wang, A.Z. Nanoparticles and their applications in cell and molecular biology. Integr. Biol. Quant. Biosci. Nano Macro 2014, 6, 9–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Diaz, D.; Care, A.; Sunna, A. Bioengineering Strategies for Protein-Based Nanoparticles. Genes 2018, 9, 370. [Google Scholar] [CrossRef] [Green Version]
  102. Liébana, S.; Drago, G. Bioconjugation and stabilisation of biomolecules in biosensors. Essays Biochem. 2016, 60, 59–68. [Google Scholar] [CrossRef] [Green Version]
  103. Boutureira, O.; Bernardes, G. Advances in chemical protein modification. Chem. Rev. 2015, 115, 2174–2195. [Google Scholar] [CrossRef] [PubMed]
  104. Scally, S.W.; McLeod, B.; Bosch, A.; Miura, K.; Liang, Q.; Carroll, S.; Reponen, S.; Nguyen, N.; Giladi, E.; Rämisch, S.; et al. Molecular definition of multiple sites of antibody inhibition of malaria transmission-blocking vaccine antigen Pfs25. Nat. Commun. 2017, 8, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Schudel, A.; Chapman, A.; Yau, M.; Higginson, C.; Francis, D.; Manspeaker, M.; Avecilla, A.; Rohner, N.; Finn, M.; Thomas, S. Programmable multistage drug delivery to lymph nodes. Nat. Nanotechnol. 2020, 15, 491–499. [Google Scholar] [CrossRef]
  106. Kwon, Y.; James, E.; Shastri, N.; Fréchet, J. In vivo targeting of dendritic cells for activation of cellular immunity using vaccine carriers based on pH-responsive microparticles. Proc. Natl. Acad. Sci. USA 2005, 102, 18264–18268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Schematic of targeted conjugate vaccines. (a) A plasmid encoding a protein subunit conjugate is transfected into mammalian cells which secrete the fusion protein for subsequent purification and administration as a vaccine. Modeled after HA targeting to Xcr1 on DCs [44]. (b) A plasmid encoding a peptide carrier and capsular PS synthases is transformed into an oligosachride transferase (OTase)—engineered strain of E. coli which assembles the glycoconjugate which is subsequently purified for use as a vaccine. Modeled after production of a Kp capsular PS-Pa exotoxin A glycoconjugate vaccine [46]. (c) NPs are chemically activated to facilitate conjugation of Ag; loaded NPs are then administered as the vaccine. Modeled after activated amines to conjugate Pfs25 and generate a malaria vaccine [47]. (d) Glycoconjugate vaccines produced by chemically activating a peptide carrier, linking the purified components, and administering the conjugate as a vaccine. Modeled after generation of the meningitis vaccine [45]. (e) A plasmid encoding a targeted protein subunit conjugate that is anchored to an OMP is transformed into bacteria which are administered as a live vaccination. The conjugate protein is trafficked to the OM and cleaved from the OMP anchor. Modeled after CR-targeted Ts Ag, produced by the engineered Salmonella vaccine strain [52]. (f) A plasmid encoding an OMP-anchored conjugate (lacking a cleavage site) is expressed in bacteria, effectively targeting the whole bacterial cell to immune cells when administered as a vaccine. Modeled after targeting Ft to CRs [51].
Figure 1. Schematic of targeted conjugate vaccines. (a) A plasmid encoding a protein subunit conjugate is transfected into mammalian cells which secrete the fusion protein for subsequent purification and administration as a vaccine. Modeled after HA targeting to Xcr1 on DCs [44]. (b) A plasmid encoding a peptide carrier and capsular PS synthases is transformed into an oligosachride transferase (OTase)—engineered strain of E. coli which assembles the glycoconjugate which is subsequently purified for use as a vaccine. Modeled after production of a Kp capsular PS-Pa exotoxin A glycoconjugate vaccine [46]. (c) NPs are chemically activated to facilitate conjugation of Ag; loaded NPs are then administered as the vaccine. Modeled after activated amines to conjugate Pfs25 and generate a malaria vaccine [47]. (d) Glycoconjugate vaccines produced by chemically activating a peptide carrier, linking the purified components, and administering the conjugate as a vaccine. Modeled after generation of the meningitis vaccine [45]. (e) A plasmid encoding a targeted protein subunit conjugate that is anchored to an OMP is transformed into bacteria which are administered as a live vaccination. The conjugate protein is trafficked to the OM and cleaved from the OMP anchor. Modeled after CR-targeted Ts Ag, produced by the engineered Salmonella vaccine strain [52]. (f) A plasmid encoding an OMP-anchored conjugate (lacking a cleavage site) is expressed in bacteria, effectively targeting the whole bacterial cell to immune cells when administered as a vaccine. Modeled after targeting Ft to CRs [51].
Molecules 26 01479 g001
Table 1. Examples of targeted therapies and vaccines.
Table 1. Examples of targeted therapies and vaccines.
Targeting UnitCargoTargeted Disease/Treatment/EffectStage of Use
Cancer Therapies
α-LIV-1 AbMonomethyl auristatin EMetastatic breast cancer [15,16,17,34]Phase 2 trials
α-HER2 AbDeruxtecanHER2-positive breast & stomach cancer [15,18]Clinical use
α-HER2 AbMertansineHER2-positive early breast cancer [23]Clinical use
α-CD33 AbN-acetyl γ calicheamicinAcute myeloid leukemia [24,25,35]Clinical use
α-CD33 AbCalicheamicinRelapsed/refractory acute myeloid leukaemia [24]Clinical use
α-CD30 AbMonomethyl auristatin EHodgkin’s lymphoma [22]Clinical use
α-CD22 AbCalicheamicinRelapsed/refractory acute lymphoblastic leukaemia [25]Clinical use
α-gp72 AbMonomethyl auristatin EModels of ovarian and colon carcinoma [30]Pre-clinical
Central Nervous System Therapies
B6 peptideCurcuminAlzheimer’s disease. Delivery of loaded NPs to TfRs on the BBB, improved memory & learning [36]Pre-clinical
TP10 peptideDopamineParkinson’s disease. Delivery of dopamine to the brain [37]Pre-clinical
LJM3064 aptamerExosomesMultiple sclerosis (EAE) associated demyelination [38]Pre-clinical
α-PSA-NECM AbSC-79Post-stroke neuro-regeneration. Delivery of SC-79 loaded NPs to neuroblasts to enhance pro-survival signaling [39]Pre-clinical
Antimicrobials
α-teichoic acid AbRifalogueEnhanced killing of intracellular MRSA [40]Pre-clinical
α toxin-reactive NPsVancomycinControlled release of antibiotic at the site of infection [41]Pre-clinical
Targeted Vaccinations
α-Clec9A AbInfluenza M2eTarget influenza Ag to DCs to enhance responses & protection [35]Pre-clinical
α-MHC-II AbInfluenza HAIncreased α-HA Ab & Th2 responses, protecting against influenza [42]Pre-clinical
α-CCR1/3/5 AbInfluenza HAIncreased CD8+ & Th1 responses, protecting against influenza [42]Pre-clinical
Xcl1Influenza HAIncreased proliferation of CD4+ & CD8+ T cells against influenza [43,44]Pre-clinical
Ccl3Influenza HATarget HA to CCR1/3/5 to induce CD4+ T cells against influenza [43]Pre-clinical
Diptheria toxoidNm PSIncreased α-Nm PS Abs, protection for meningococcal disease [45]Clinical use
Pa exotoxin protein AKp PSIncreased α-Kp PS Abs, protection against Kp infection [46]Pre-clinical
Qβ VLPsPfs25Increased transmission-blocking Abs against malaria [47]Pre-clinical
MannosehepB DNAAPC transfection via MR, stimulating α-HepB responses [48]Pre-clinical
C3dSe FimAIncreased immunogenicity of FimA, protection against Se [49]Pre-clinical
C3dHIV1 EnvIncreased neutralizing Ab production against HIV1 [50]Pre-clinical
C3dFt whole cellsIncreased Ag binding to APCs, protection against tularemia [51]Pre-clinical
C3d p28Ts Ag30Increased Ab production, protection against trichinosis [52]Pre-clinical
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Tummillo, K.M.; Hazlett, K.R.O. Co-Opting Host Receptors for Targeted Delivery of Bioconjugates—From Drugs to Bugs. Molecules 2021, 26, 1479. https://doi.org/10.3390/molecules26051479

AMA Style

Tummillo KM, Hazlett KRO. Co-Opting Host Receptors for Targeted Delivery of Bioconjugates—From Drugs to Bugs. Molecules. 2021; 26(5):1479. https://doi.org/10.3390/molecules26051479

Chicago/Turabian Style

Tummillo, Kristen M., and Karsten R.O. Hazlett. 2021. "Co-Opting Host Receptors for Targeted Delivery of Bioconjugates—From Drugs to Bugs" Molecules 26, no. 5: 1479. https://doi.org/10.3390/molecules26051479

Article Metrics

Back to TopTop