Next Article in Journal
3D Printing of Drug-Loaded Thermoplastic Polyurethane Meshes: A Potential Material for Soft Tissue Reinforcement in Vaginal Surgery
Next Article in Special Issue
Skin Irritation Testing beyond Tissue Viability: Fucoxanthin Effects on Inflammation, Homeostasis, and Metabolism
Previous Article in Journal
Energy Transfer in Vials Nested in a Rack System During Lyophilization
Previous Article in Special Issue
Amphiphilic Polypeptides for VEGF siRNA Delivery into Retinal Epithelial Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Antibodies for the Treatment of Brain Metastases, a Dream or a Reality?

Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
*
Authors to whom correspondence should be addressed.
Pharmaceutics 2020, 12(1), 62; https://doi.org/10.3390/pharmaceutics12010062
Submission received: 19 November 2019 / Revised: 13 December 2019 / Accepted: 28 December 2019 / Published: 13 January 2020
(This article belongs to the Special Issue Drug Delivery across Biological Barriers)

Abstract

:
The incidence of brain metastases (BM) in cancer patients is increasing. After diagnosis, overall survival (OS) is poor, elicited by the lack of an effective treatment. Monoclonal antibody (mAb)-based therapy has achieved remarkable success in treating both hematologic and non-central-nervous system (CNS) tumors due to their inherent targeting specificity. However, the use of mAbs in the treatment of CNS tumors is restricted by the blood–brain barrier (BBB) that hinders the delivery of either small-molecules drugs (sMDs) or therapeutic proteins (TPs). To overcome this limitation, active research is focused on the development of strategies to deliver TPs and increase their concentration in the brain. Yet, their molecular weight and hydrophilic nature turn this task into a challenge. The use of BBB peptide shuttles is an elegant strategy. They explore either receptor-mediated transcytosis (RMT) or adsorptive-mediated transcytosis (AMT) to cross the BBB. The latter is preferable since it avoids enzymatic degradation, receptor saturation, and competition with natural receptor substrates, which reduces adverse events. Therefore, the combination of mAbs properties (e.g., selectivity and long half-life) with BBB peptide shuttles (e.g., BBB translocation and delivery into the brain) turns the therapeutic conjugate in a valid approach to safely overcome the BBB and efficiently eliminate metastatic brain cells.

Graphical Abstract

1. Brain Metastases

Brain metastases (BM) account for significant morbidity and mortality. The exact incidence is unknown [1,2]. Based on various studies, investigators estimate that BM occurs in 10%–20% of adult patients with cancer [3]. Nevertheless, the incidence might be higher, and it is increasing due to prolonged life expectancy, increased resistance to cancer therapies, and improved imaging techniques. In addition, the increased patient survival by treating primary tumors may increase the number of patients that will develop more aggressive BM, or that are resistant to therapy. Among the different cancer types, lung cancer (19.9%), breast cancer (15.2%), and melanoma (6.9%) are the most common primary tumors developing BM [4]. After diagnosis, overall survival (OS) is poor. However, early diagnosis, improved systemic therapies, and multimodality treatments have significantly increased patients’ survival [5].

1.1. BM Pathophysiology

The pathophysiology of BM is complex and involves a multi-step process constituted of two major stages (Figure 1) [6]. The first stage is tumor migration, which includes (i) metastatic clone progression, due to tumor cells’ ability to degrade extracellular matrix (ECM); (ii) intravasation (transendothelial migration of cancer cells into vessels); (iii) dissemination (spread of tumor cells via bloodstream); (iv) extravasation (transendothelial migration of cancer cells into tissues). The second stage corresponds to tumor colonization.
The cells presented in the primary tumor are heterogeneous. Among others, the tumor microenvironment is composed of cancer stem cells (CSCs), partially differentiated progenitor cells, and fully differentiated end-stage cells [6]. Recent findings attribute to CSCs the primary responsibility for enhanced malignancy since they can complete the two stages of metastases formation (Figure 1) [7]. However, during cancer progression, other cells undergo an epithelial–mesenchymal transition (EMT), changing their plasticity by morphological and phenotypical conversions [8,9]. EMT enables non-CSCs to resemble a CSC state. Thus, they acquire the ability to invade and colonize distant sites, creating secondary niches that may progress to a secondary tumor [10]. Therefore, in the end, within the tumor microenvironment, all cells are malignant. Nevertheless, the development of distal metastases only occurs in <0.1% of disseminated cancer cells. Thus, although the formation of metastases represents a major threat, it is considered highly inefficient [8,11].

1.2. BBB Physiology

BBB is a complex system composed of a structurally distinct and continuous endothelial cell layer separating two brain compartments, namely, the blood and extracellular fluid. Its components include an endothelial cell layer, adjoined by tight cell-to-cell junction proteins, and pinocytic vesicles [12]. All together, they contribute to the selective permeability of the barrier, allowing brain homeostasis. The BBB is also dynamic. It responds to regulatory signals from both the blood and the brain [13], being the main portal into the brain of gaseous molecules, such as O2 and CO2, ions, nutrients, hormones, and water (Figure 2). Hydrophobic compounds (<500 Da) diffuse across the endothelium membrane. Carrier-mediated transport (CMT) is responsible for the transport of glucose and amino acid residues. While water-soluble molecules (e.g., ions) cross the BBB through ion channels. On the other hand, macromolecules (proteins and peptides) transport rely on endocytic vesicles, which involve either receptor-mediated transport (RMT) or adsorptive-mediated transport (AMT) [14,15].
Nevertheless, the BBB interacts with the metastatic cells in an unidentified way. The BBB is hypothesized to create a unique brain microenvironment and to influence metastatic colonization [16]. The increased permeability of tumor-associated endothelial cells, due to tumor penetration into the brain, permits leakage of proteins and water into brain parenchyma. The mechanism described is responsible for the edema often associated with BM [17]. Microglia and macrophages influence tumor proliferation and invasion by secreting multiple cytokines, growth factors, enzymes, and reactive oxygen species (ROS). Other immune cells may also participate in the BBB translocation. However, the exact mechanisms are debatable [18,19,20,21]. The BBB structure may be affected momentarily during cancer cells’ invasion; however, in other non-cancer-related central-nervous system (CNS) pathologies, only in advanced disease stages, the dysfunction is usually significant.

1.3. BM Treatment

Major developments have been made in understanding brain function, metastases progression, and the development of medical technologies. However, in many cases, the major drawback in BM treatment is the inefficient drug delivery into the brain [22,23]. The BBB remains the most significant obstacle to the efficient delivery of small-molecule drugs (sMDs) and therapeutic proteins (TPs) [24]. In addition, some authors also attributed to the therapeutic resistance of metastatic cells the responsivity for therapy inefficiency. According to them, during cancer progression, some survival pathways, such as the PI3K/AKT/mTOR are activated in specific cells, which contributes to the poor response of metastatic cells [25,26]. In the end, both mechanisms might be contributing to therapeutic failure.
The first-line approach to treat BM includes surgery, stereotactic radiosurgery (SRS), and whole-brain radiation therapy (WBRT) [27,28,29]. However, both systemic and intracranial disease control are also possible with the improved systemic therapies that have begun to offer greater potential for specific cancer types and genotypes. Thus, the management of BM has become increasingly individualized [1]. Depending on the histology and systemic disease status, physicians may consider all the available therapies. For instance, recommendations suggest better disease management by using a multidisciplinary modality in patients with BM from breast cancer, melanoma, and specific genotypes of non-small cell lung cancer (NSCLC) (e.g., epidermal growth factor receptor (EGFR) gene mutations, translocations in the anaplastic lymphoma kinase (ALK) gene). Whenever possible, patients may enroll in clinical trials for a novel or existing therapy. Still, given the paucity of effective treatment options, BM elimination represents an unmet clinical need [30].

2. Strategies to Overcome the BBB

The BBB restricts the delivery of therapeutics to the brain. Overall, 98% of sMDs and probably all TPs cannot cross the barrier by free diffusion [31,32,33]. In the last decade, intense investigation allowed the discovery of new strategies to increase brain penetration of existing therapeutics (invasive, pharmacological, and physiological) [34,35,36]. Ideally, the translocation should not compromise the BBB integrity. However, some of the current strategies do not meet this criterion, namely, the invasive and pharmacological approaches.

2.1. Invasive Approach

This strategy allows drugs to flow directly from the systemic circulation into the brain by BBB disruption using different methodologies. The most important are: (1) osmotic disruption, due to the administration of hypertonic solutions (e.g., mannitol) causing cells shrinking based on cell dehydration [37]; (2) ultrasound methods, which rely on transcranial delivery of low-frequency ultrasound waves resulting in the opening of tight junctions [38]; and (3) pharmacological agents, such as bradykinin-like compounds (e.g., histamine, bradykinin) that disrupt tight junctions by stimulating B2 receptors presented in endothelial cells and transiently increasing cytosolic Ca2+ [39]. The costs, anesthetic administration, and hospitalization are significant drawbacks for all these approaches. Also, the disruption of the BBB may increase tumor dissemination, as well as irreversible neuropathological changes due to the entry of unwanted substances [34].

2.2. Pharmacological Approach

The pharmacological approach relies on the observation that some molecules freely enter the brain owing to their molecular weight (<500 Da), charge (low hydrogen bonding capabilities), and lipophilicity [40]. Thus, researchers started modifying, through medicinal chemistry, molecules that are active against CNS diseases or BM to enable them to get into the brain [31]. Although it has enormous potential, the modifications may result in loss of pharmacological activity. In addition, the new molecule may become a substrate for the efflux pumps by increasing drugs’ lipophilicity, which decreases brain accumulation [34].

2.3. Physiological Approach

This natural strategy exploits the various transporters and receptors expressed at the BBB, as well as the physiological properties of the BBB (e.g., charge and lipid composition) (Figure 2) [41,42,43]. These translocation mechanisms are fundamental for the uptake of essential substances to maintain brain homeostasis. They can be classified into: (1) CMT, which are responsible for the cross of glucose (glucose transporter—GLUT1), amino acids (large neutral amino acid transporter—LAT1, and cationic amino acid transporter—CAT1), and nucleosides (nucleobase transporter—NBT); (2) RMT, fundamental for large molecules translocation, such as transferrin (transferrin receptor—TfR), insulin (insulin receptor—IR), low-density lipoprotein (lipoprotein receptor-mediated protein—LRP), leptin (leptin receptor—LEPR), and fragment crystallizable (Fc) fragment of immunoglobulin G (IgG) (Fc fragment of IgG receptor transporter α—FCGRT); and finally, (3) AMT that drives albumin and other plasma proteins to brain [44].
CMT is an interesting transporter due to the easy coupling of endogenous substrates to sMDs [45]. Besides, it is also possible to perform direct modification of sMDs to resemble CMT natural substrates. The changes allow drugs to be recognized and transported across the BBB [46]. Nevertheless, the molecules generated: (1) must mimic that of the endogenous CMT substrate; (2) should not affect CMT physiological function; and (3) must maintain its pharmacological activity. So far, targeting nanocarriers to CMTs have been the best example of the strategy’s success. However, the application of this approach was only possible for small molecules, as revised in Witt et al. [47].
Another promising strategy to develop molecules that can efficiently cross the BBB is the RMT. These molecules are known as Trojan Horses and can be either peptides or antibodies [48]. TfR and IR are the most important BBB receptors explored by researchers. Pardridge et al. have extensively documented the use of antibodies targeting these receptors [49,50,51]. The in vivo studies demonstrated an accumulation of different anti-TfR monoclonal antibodies (mAbs) in the brain tissue and a distinct biodistribution. The high affinity of antibodies towards these receptors is, however, a limitation since results in weak receptor dissociation. Consequently, the high-affinity antibodies follow the lysosomes pathway during intracellular trafficking leading to its degradation [52].
Yu et al. elegantly solved the problem by reducing the affinity of anti-TfR mAbs [53]. Next, the group developed a bispecific therapeutic antibody with a low affinity for TfR and a high affinity for the enzyme β-secretase (BACE1), an Alzheimer’s disease drug target. Relevant results were obtained from the evaluation of the bispecific antibody efficacy in non-human primates. The brain accumulation was significantly higher than control, and the amyloid β-peptide presence in the brain and serum reduced considerably [54]. Similarly to TfR, exciting studies targeting IR have been developed. Pardridge et al. have shown a total of 4% brain uptake 3 h after intravenous administration in Rhesus monkeys. In the treatment of Parkinson’s, stroke, metachromatic leukodystrophy, and Sanfilippo type A syndrome, some therapeutic drugs have been linked to the mAb and successfully translocated across the BBB [49,55].
Another interesting strategy to deliver drugs into the brain exploiting RMT is the use of nanoparticles (NPs) coupled with mAbs or peptides that recognize these receptors [43,56,57]. NPs are colloidal carriers of natural or synthetic origin with a size varying from 1 to 1000 nm. They are a fascinating system due to their modulating capacity concerning shape, size, hydrophobicity, coating, chemistry, and surface charge [58]. In addition, they also have a high capacity of drug payload, the relatively few mAbs or peptides to achieve high levels of drug targeting, protection of the encapsulated drug, and the ability to provide a controlled release of the drug [59,60].
Although the considerable achievements accomplished, the drawback of these RMT systems is related to the competition with natural substrates, which may affect brain homeostasis; and may result in receptors’ saturation due to the high affinity of antibodies [34,61,62]. To overcome these limitations, recently, more attention has been given to AMT. The concept of AMT through the BBB began with the observation that polycationic proteins’ brain uptake did not involve binding to the endothelial cell surface [63]. Electrostatic interaction between positively charged substances and negatively charged BBB drives the translocation. The vesicles created allow BBB cross and, consequently, brain accumulation [64]. Lack of selectivity of these systems and possible BBB disruption were the major concerns highlighted by researchers. However, the recent proof-of-concept given by the use of cell-penetrating peptides (CPPs) in BBB translocation (BBB peptide-shuttle) has launched a new interest in this strategy. These peptides have demonstrated a natural selectivity towards negatively charged membranes and the ability to translocate large cargoes without BBB damage both in vitro and in vivo models [65,66,67]. Therefore, the optimization of BBB peptide shuttles based-systems in the delivery of sMDs and TPs, using AMT, will be an area of intense investigation during the next decade.

3. Therapeutic Antibodies for BM Elimination

TPs are the standard of care in a number of therapeutic areas [68]. They are protein manufactured for biopharmaceutical use and include, for instance, mAbs, peptides, growth factors, cytokines, and enzymes [69]. Their production is relatively easy and relies mostly on either simple purification or recombinant DNA technology. Throughout the following sections, the description of antibodies’ activity and their therapeutic value only concern human antibodies. The activity of antibodies is species dependent. Thus, some features presented might not be accurate for non-human antibodies.

3.1. Monoclonal Antibodies

mAbs represent the fastest growing class of TPs. Currently, over 50 therapeutic antibodies are on the market [70]. They are complex molecules consisting of homodimers of variable and constant regions (Figure 3) [71]. The former has antigen specificity owing to the presence of complementary determining regions (CDRs). On the other hand, the Fc domain is responsible for the long half-life of antibodies, due to antibody recycling after interaction to the neonatal Fc receptor (FcRn) [72]; and immune activation (complement-dependent cytotoxicity—CDC; and antibody-dependent cellular cytotoxicity—ADCC) by engaging Fcγ receptors (FcγRs) on immune cells (e.g., neutrophils, natural killer cells, monocytes) [73]. High therapeutic tolerability and low risk-to-benefit ratios favor the use of therapeutic antibodies. Thus, their exquisite specificity, high binding affinity, long half-life, low toxicity, and versatility are characteristics that contributed to antibodies’ success [74,75]. Additionally, the low number of drug–drug interactions between mAbs and sMDs increased their combination in many therapeutic regimens [70].
The mechanism of action of mAbs differs depending on the molecule engineered (Figure 3). They can target soluble mediators (e.g., cytokines) to inhibit their binding to receptors and, consequently, inhibit signaling; or they can target membrane receptors either inducing or antagonizing signaling (e.g., programmed-cell death ligant-1—PD-L1; or human epidermal receptor-2—HER2, respectively) [76]. In addition, the presence of the Fc domain allows immune stimulation (CDC and/or ADCC). CDC is related to complement activation. Complement is one of the first mediators of the immune response to pathogens and cells. After binding, antibodies activate the classical complement cascade. Thus, releasing cytokines (e.g., anaphylatoxins and opsonins) and forming the membrane attack complex (MAC), which lead to cell lysis and phagocytosis [77]. On the other hand, ADCC occurred due to the interaction of the Fc domain with FcγRs on effector immune cells (e.g., neutrophils—FcγRI, natural killer cells—FcγIIIA; or monocytes—FcγIIIB). After recognition of an antibody-coated target cell, effector cells engage the release of granzymes and perforins [78]. The consequence is cell death. The magnitude of the stimulation of either CDC or ADCC depends on the IgG subset (IgG 1–4). For instance, IgG2 and IgG4 do not activate both mechanisms. Therefore, they are designed primarily for signaling blockage. Oppositely, IgG1 and IgG3 strongly activate both CDC and ADCC. Owing to its short half-life due to a low FcRn affinity, IgG3 does not have the therapeutic value of other IgG subsets [79].

3.2. Therapeutic Value

The first mAb approved was muromonab-CD3 in the prevention of transplant rejection. Ever since, mAbs have been introduced in a number of therapeutic regimens in a wide range of conditions, such as organ transplantation (e.g., basiliximab and belatacept), inflammatory diseases (e.g., adalimumab and tocilizumab), and cancer (e.g., trastuzumab and cetuximab) [70]. The use of antibodies is increasing and improved mAb-based strategies will appear on the market in response to current therapeutic challenges (Figure 4). In particular, antibody research focused on the development of antibody fragments (e.g., single-chain Fv—scFv, single-domain antibody—sdAb, antigen-binding fragments—Fab); antibody–drug conjugates (ADC) (e.g., trastuzumab emtansine); fusion proteins (e.g., etanercept); and intrabodies [80,81,82]. Although physicians use mAbs in a variety of conditions, their applicability in the treatment of CNS diseases and BM remains challenging. Nevertheless, there are some mAb-based systems already approved (Table 1) or in investigation (Table 2).

3.3. CNS Diseases

Multiple sclerosis and episodic headache are the only neurologic pathologies where mAbs have been administrated, which started with the approval of natalizumab [83] and erenumab [84], respectively. Natalizumab is an IgG4 mAb targeting α4β1-integrin (very late activation antigen-4—VLA-4), which is present on the surface of leukocytes. After binding to VLA-4, the mAb inhibits the interaction between VLA-4 and vascular cell adhesion molecule-1 (VCAM-1). Consequently, reducing the adhesion, attachment, and migration of leukocytes across the BBB into the CNS [85]. In a pivotal phase III trial (AFFIRM), natalizumab reduced clinical relapse at one-year by 68% and the risk of continuous progression of disability by 42–54% over two years [86]. Nevertheless, the therapeutic effect observed occurs due to a peripheral action, instead of a direct antibody penetration into the brain. Erenumab is an IgG2, which targets the calcitonin gene-related peptide (CGRP) receptor [87]. The mAb competes with the binding of CGRP and inhibits its function at the CGRP receptor. The CGRP receptors are located at relevant sites to migraine pathophysiology, such as the trigeminal ganglion and the paraventricular structures. The BBB does not protect these regions. Thus, erenumab also exerts action at the periphery and not at the brain. In phase III STRIVE clinical trial, erenumab was able to significantly reduce the number of migraine days per month by 3.2 versus 1.8 in the placebo group. Efficacy was sustained up to one year [88].
A different strategy was applied in the management of Alzheimer’s disease. In this case, instead of antibodies acting at the periphery, researchers are using mAbs and re-engineered antibody fragments targeting natural brain portals (e.g., TfR and IR) [44]. Pardridge et al. reported for the first time an anti-β-amyloid (Aβ) scFv fused to mAbs targeting either TfR or IR [89]. The brain uptake of the molecule was of 0.88% ID/brain. The value is within the boundaries of the brain uptake of other drugs that are active in the brain [44,90,91]. However, the mAb demonstrated a high affinity towards the receptor. Thus, receptor dissociation was a major limitation. The consequence might be antibody degradation since the molecules, instead of crossing the BBB, follow the lysosomal pathway. Yu et al. elegantly solved the problem by reducing the affinity of these mAb [53]. In addition, the group further developed a bispecific therapeutic antibody with a low affinity for TfR and a high affinity for another Alzheimer’s disease drug target, the BACE1 [54]. The brain accumulation of the molecule in non-human primates was significantly higher than control, and the amyloid β-peptide presence in the brain and serum reduced considerably. The drawback of these RMT systems is the competition with natural substrates, which may affect brain homeostasis; and receptor saturation due to the high affinity of the antibodies engineered [44].

3.4. Brain Metastases

In the treatment of BM, the reality is different. In the recruitment phase of clinical trials, an active exclusion of patients presenting BM occurs [92,93]. Therefore, to date, no clinical trial supports the use of mAbs in the management of BM. The lack of information concerning efficacy and safety are the main reasons. Consequently, the standard cancer regimens with well-established antibody-based treatments cannot be applied to patients with BM [94]. Their use by physicians represents an off-label use. Nevertheless, a class of mAbs related to immunotherapy (e.g., nivolumab, pembrolizumab) is showing promising results. Several studies suggest their role in the elimination of metastatic tumors, such as in the brain. Still, more data is necessary to approve these antibodies in the treatment of metastatic brain cancers [95]. Another promising field is the radionuclide therapy. In the last years, numerous papers have been published reporting its success in brain tumors and metastases [96].
New therapeutic targets in metastatic progression in either primary or secondary tumors have driven intense research into the development of mAb-based systems [97,98], as they offer effective targeted treatment with low adverse events. However, the lack of specificity and poor BBB penetration render them ineffective. It is therefore imperative to find strategies that allow antibody translocation across the BBB. For instance, the use of antibody fragments to reduce their molecular weight. Or modify the mAbs to contain a translocation moiety, such as a CPPs. CPPs are effective in the delivery of large cargoes across cell membranes and even across the BBB (BBB peptide shuttles) [66,99]. Similar to the Trojan horse approach they engage interaction with BECs and BBB translocation, the main advantage being that CPP does not require receptors in the majority of the cases, thus reducing the toxicity of the system significantly.

4. The Role of BBB Peptide-Shuttles

CPPs are short peptides (less than 30 amino residues) capable of crossing cell membranes without causing significant membrane damage [99]. They represent a broad group of peptides with different physicochemical properties. Accordingly, they can be: (1) cationic, which comprises peptides with highly positive charges at physiological pH; (2) amphipathic, that contains both polar (hydrophilic) and nonpolar (hydrophobic) regions of amino acids; and (3) hydrophobic, the less studied class, which are CPPs mainly containing nonpolar residues, resulting in a low net charge. Naturally occurring proteins and peptides are the principal sources of CPPs [105,106], however, to optimize the peptides’ properties, fully engineered peptides have been designed, based on computational modeling [107].
The specific internalization mechanism of CPPs is unclear [108,109]. The peptide’s concentration, the cargo conjugated, the physicochemical properties, and molecular weight are features affecting the efficiency of cellular entry, as well as the internalization pathway followed. Nevertheless, energy-dependent endocytic pathways, which include clathrin-mediated endocytosis, caveolin-mediated endocytosis, and macropinocytosis, are considered the main translocation mechanisms [110]. The intensive research using peptides and the development of technologies that allowed their conjugation to TPs (e.g., recombinant DNA technology) resulted in the capacity of cargo-transportation not only across cell membranes, especially epithelia, but also the endothelial BBB (BBB peptide shuttle).
Human Immunodeficiency Virus Trans-activator of transduction (TAT) peptide was the first peptide demonstrating translocation properties [111]. Subsequently, many other peptides, like SynB, Penetratin, Angiopep-2, dNP2, and PepH3 were studied with relevant results [112]. Despite the variation in length and amino acids’ sequence, these peptides share common features. Among others, their amphipathic nature, net positive charge, theoretical hydrophobicity, helical moment, as well as the ability to interact with lipid membranes. The mechanism by which these peptide shuttles cross the BBB and mediate cargo translocation is not fully understood and may vary according to the concentration, cell type, and the cargo of interest [113]. Direct membrane permeation, RMT, and AMT are the three principal possibilities. The latter constitutes an advantage compared with others since it avoids enzymatic degradation, problems related to endosomal escape, receptor saturation, and toxicity, among others. The process is based on the electrostatic binding of positive charge peptide-shuttle to negative charge proteoglycans (Figure 2), forming a vesicle that transports the system across the endothelial cells layer [112].
The efficiency of large proteins delivery, such as antibodies or fusion proteins across cell membranes by CPPs, has been intensively studied, mainly in vitro. However, the delivery to the brain by peptide shuttles was not. Schwarze et al. performed the first in vivo study using BBB peptide shuttles. In their work, they successfully delivered a 120 kDa β-galactosidase fused to TAT into the brain [114]. These results showed that the direct delivery of proteins into the brain was possible. Afterward, others conjugated TAT to B-cell lymphoma-extra-large (TAT-Bcl-xL), glial cell-derived neurotrophic factor (TAT-GDNF), NR2B9c (TAT-NR2B9c), and c-Jun N-terminal kinase-1 (TAT-JNK1) fusion proteins, and evaluated their concentration in the brain [115]. In addition, the use of rabies virus glycoprotein (RVG) fused to brain-derived neurotrophic factor (RVG-BDNF), and fibroblast growth factor-4 (FGF4) fused to suppressor of cytokine signaling-3 (FGF4-SOCS3) also validated the use of these peptide shuttles [116,117].
Angiopep-2 is a 19-amino acid peptide, derived from the Kunitz domain, which binds to LRP1 and efficiently penetrates the BBB via RMT. In the study performed by Demeule et al., the translocation of angiopep-2 in an in vitro BBB model was found to be seven-fold higher than of aprotinin, an LRP1 natural ligand with BBB translocation properties [118]. Furthermore, the apparent distribution of the peptide shuttle in vivo was far greater than both transferrin and aprotinin, confirming the BBB translocation capabilities of angiopep-2. To further challenge the peptide, researchers conjugated it to an anti-HER2 mAb to investigate the ability of cargo translocation across the BBB. HER2+ breast cancer patients demonstrate a high incidence of BM. The low concentration of mAb in the brain provides a “sanctuary site” for tumor proliferation. Nevertheless, in this study, after carotid artery administration, 60% of the molecule was localized in the brain, demonstrating high brain accumulation. Besides, increased survival was reported compared with control [119].
dNP2 is an amphiphilic human-derived CNS-permeable peptide shuttle. To evaluate the abilities of cargo translocation across the BBB, Lim et al. conjugated the peptide with the cytoplasmic domain of CTLA4 (ctCTLA-4) [120]. CTLA-4 is an immune regulatory receptor expressed on the surface of T cells and often associated with susceptibility to multiple sclerosis. Thus, proving the autoimmune pathogenesis of the disease. In Lim et al. study, the administration of the dNP2-conjugated ctCTLA-4 protein successfully controlled autoimmune effector T-cell responses in an experimental autoimmune encephalomyelitis (EAE) model, an experimental mouse model of multiple sclerosis. The exact mechanism of BBB translocation and cell internalization were not determined. However, due to dNP2 properties, BBB crossing was considered to be AMT, and cellular uptake of the fusion protein through lipid-raft mediated endocytosis [120].
The most recent peptide shuttle reported is PepH3, a cationic peptide derived from Dengue virus type-2 capsid protein (DEN2C) [121]. In Neves et al. study, the in vitro BBB transmigration after 24 h was 67.2%. Furthermore, in an in vivo model, the peptide showed a brain biodistribution of 0.31% after 5 minutes. Although the exact mechanism is not fully described, studies with endocytosis inhibitors reveal that the PepH3 mechanism is consistent with the AMT. The peptide BBB translocation capacity was also evaluated in conjugation to anti-β-amyloid protein 42 (bAP42) sdAb (anti-bAP42 sdAb) [122]. bAP42 is an amyloid precursor protein fragment that plays a significant role in the formation of “senile plaques” characteristic of Alzheimer’s disease. Through conjugation of PepH3 to an antibody fragment, the investigators expected the increase in antibody concentration in the brain, followed by binding to bAP42, decreasing plaques formation. Also, the complex may be detected in peripheral blood aiding in the diagnosis of disease. Interestingly, the PepH3-anti-bAP42 sdAb conjugate showed a brain accumulation of 1.5% after the same period (5 minutes), showing that cargoes do influence the translocation mechanisms of BBB peptide shuttles [122].
The results obtained with these fusion proteins are promising. Nevertheless, their high elimination rates prevent the clinical use of such peptide shuttles-based systems. Peptides and small proteins are rapidly metabolized by serum/tissue proteases and easily eliminated by glomerular filtration [123]. Since they do not present a high circulation time, BBB translocation is also affected. By increasing circulation time, the brain concentration the conjugates will improve, due to a higher contact frequency and interaction time. Thus, increasing the half-life of such systems will hypothetically augment brain accumulation. Several strategies are available. The most interesting methods explore the human serum albumin protein (HSA) and the FcRn mediated recycling [124].
In the peptide shuttles-based approaches mentioned, researchers used mainly antibody fragments of a full IgG as therapeutic agents [120,122]. However, antibodies are proteins also used as pharmacokinetic enhancers owing to their Fc region. Thus, a strategy combining these properties with the translocation capabilities of peptide shuttles will improve the half-life of the system and increase brain accumulation. Depending on the target selected, the strategy may be used to eliminate BM or CNS diseases. Since antibodies are large proteins and BBB translocation is complicated, instead of a full antibody, the engineering of a minibody comprising the antibodies’ minimal domains (Fc fragment and variable region) to keep the antibody properties is preferable. In this, a reduction from 150 kDa for 25–80 kDa is achieved (Figure 4). These molecular weights are similar to the fusion proteins studied, increasing the interest of the strategy proposed. Another approach is the nanobodies (smallest Ab-derivative) which reveal promising results in preclinical and clinical studies in the elimination of brain tumors [102,125,126].

5. Conclusions and Future Perspectives

Our understanding of cancer biology and technological advances in cancer diagnosis and therapy improved significantly over the past decades. One of the landmarks for reversing the worldwide increase in cancer incidence and mortality was the development of more effective, tumor-specific, and less toxic anti-cancer drugs. The use of targeted therapy of human cancers using mAb-based systems has revolutionized cancer therapy. They are currently being used as the first choice to treat some of the most frequent metastatic cancers, such as HER2+ breast cancers or colorectal cancers. More recently, the efficacy demonstrated by antibodies inhibiting immune checkpoints has extended their use in other tumor types. In addition, they have also been introduced in many therapeutic protocols in combination with sMDs. For the treatment of cancer, the Food and Drug Administration (FDA) and the European Medicines Agency (EMA) have approved 32 mAbs-based systems. Interestingly, between 2012 and 2017 the number has doubled. Therefore, the value of mAbs in cancer therapy is undisputable.
Despite the excitement of mAbs-based systems in cancer treatment, their use in brain cancers and BMs are limited since they are unable to cross the BBB. Their molecular weight and hydrophilic nature difficult brain accumulation. Therefore, to create BM specific mAbs-based systems, they need to be modified to get across the BBB. Many strategies have been employed for mAbs to become a reality in the treatment of BMs. The most promising uses BBB peptide shuttles conjugated to mAbs-based systems. These peptides use endogenous routes, such as RMT or AMT to cross the BBB. Although RMT has been by far the most exploited route, it presents several disadvantages, namely, the receptor’s saturation and natural ligand competition. Therefore, with the advent of many peptide shuttles exploring AMT, the crossing of the BBB without interfering with brain homeostasis has become a reality. Consequently, the number of studies involving BBB peptide shuttles mAb-based systems are increasing in the literature.
In conclusion, BBB peptide shuttles mAb-based systems are being designed and studied with some limitations. However, the attractive results within different studies validate their application. Consequently, in the near future, it is expected a significant increase in the number of molecules conjugated to BBB peptide shuttles pushing the use of antibodies for the treatment of BMs into a reality.

Author Contributions

M.C., D.G., M.A.R.B.C., and V.N. contributed to the structuring of the present review, to the literature search, and to the writing of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Portuguese Funding Agency, Fundação para a Ciência e Tecnologia, FCT IP, grants PD/BD/128281/2017, PTDC/BBB-BQB/1693/2014 and PTDC/BBB-NAN/1578/2014.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lin, X.; DeAngelis, L.M. Treatment of Brain Metastases. J. Clin. Oncol. 2015, 33, 3475–3484. [Google Scholar] [CrossRef] [PubMed]
  2. Cagney, D.N.; Martin, A.M.; Catalano, P.J.; Redig, A.J.; Lin, N.U.; Lee, E.Q.; Wen, P.Y.; Dunn, I.F.; Bi, W.L.; Weiss, S.E.; et al. Incidence and prognosis of patients with brain metastases at diagnosis of systemic malignancy: A population-based study. Neuro-Oncology 2017, 19, 1511–1521. [Google Scholar] [CrossRef] [Green Version]
  3. Nayak, L.; Lee, E.Q.; Wen, P.Y. Epidemiology of Brain Metastases. Curr. Oncol. Rep. 2012, 14, 48–54. [Google Scholar] [CrossRef] [PubMed]
  4. Tabouret, E.; Chinot, O.; Metellus, P.; Tallet, A.; Viens, P.; GonÇAlves, A. Recent Trends in Epidemiology of Brain Metastases: An Overview. Anticancer Res. 2012, 32, 4655–4662. [Google Scholar] [PubMed]
  5. Eichler, A.F.; Loeffler, J.S. Multidisciplinary Management of Brain Metastases. Oncology 2007, 12, 884–898. [Google Scholar] [CrossRef] [PubMed]
  6. Massagué, J.; Obenauf, A.C. Metastatic Colonization. Nature 2016, 529, 298–306. [Google Scholar] [CrossRef]
  7. Ren, D.; Zhu, X.; Kong, R.; Zhao, Z.; Sheng, J.; Wang, J.; Xu, X.; Liu, J.; Cui, K.; Zhang, X.H.F.; et al. Targeting Brain-Adaptive Cancer Stem Cells Prohibits Brain Metastatic Colonization of Triple-Negative Breast Cancer. Cancer Res. 2018, 78, 2052–2064. [Google Scholar] [CrossRef] [Green Version]
  8. van Zijl, F.; Krupitza, G.; Mikulits, W. Initial steps of metastasis: Cell invasion and endothelial transmigration. Mutat. Res. 2011, 728, 23–34. [Google Scholar] [CrossRef]
  9. Jeevan, D.S.; Cooper, J.B.; Braun, A.; Murali, R.A.J.; Jhanwar-Uniyal, M. Molecular Pathways Mediating Metastases to the Brain via Epithelial-to-Mesenchymal Transition: Genes, Proteins, and Functional Analysis. Anticancer Res. 2016, 36, 523–532. [Google Scholar]
  10. Mani, S.A.; Guo, W.; Liao, M.-J.; Eaton, E.N.; Ayyanan, A.; Zhou, A.Y.; Brooks, M.; Reinhard, F.; Zhang, C.C.; Shipitsin, M.; et al. The epithelial–mesenchymal transition generates cells with properties of stem cells. Cell 2008, 133, 704–715. [Google Scholar] [CrossRef] [Green Version]
  11. Celià-Terrassa, T.; Kang, Y. Metastatic niche functions and therapeutic opportunities. Nat. Cell Biol. 2018, 20, 868–877. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Serlin, Y.; Shelef, I.; Knyazer, B.; Friedman, A. Anatomy and Physiology of the Blood–brain Barrier. Semin. Cell Dev. Biol. 2015, 38, 2–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Strazielle, N.; Ghersi-Egea, J.F. Physiology of Blood–Brain Interfaces in Relation to Brain Disposition of Small Compounds and Macromolecules. Mol. Pharm. 2013, 10, 1473–1491. [Google Scholar] [CrossRef] [PubMed]
  14. Sharif, Y.; Jumah, F.; Coplan, L.; Krosser, A.; Sharif, K.; Tubbs, R.S. Blood brain barrier: A review of its anatomy and physiology in health and disease. Clin. Anat. 2018, 31, 812–823. [Google Scholar] [CrossRef]
  15. Daneman, R.; Prat, A. The blood–brain barrier. Cold Spring Harb. Perspect. Biol. 2015, 7, a020412. [Google Scholar] [CrossRef] [Green Version]
  16. Weil, R.J.; Palmieri, D.C.; Bronder, J.L.; Stark, A.M.; Steeg, P.S. Breast Cancer Metastasis to the Central Nervous System. Am. J. Pathol. 2005, 167, 913–920. [Google Scholar] [CrossRef] [Green Version]
  17. Lesniak, M.S.; Brem, H. Targeted therapy for brain tumours. Nat. Rev. Drug Discov. 2004, 3, 499–508. [Google Scholar] [CrossRef]
  18. Green, C.E.; Liu, T.; Montel, V.; Hsiao, G.; Lester, R.D.; Subramaniam, S.; Gonias, S.L.; Klemke, R.L. Chemoattractant Signaling between Tumor Cells and Macrophages Regulates Cancer Cell Migration, Metastasis and Neovascularization. PLoS ONE 2009, 4, e6713. [Google Scholar] [CrossRef] [Green Version]
  19. Gonzalez, H.; Hagerling, C.; Werb, Z. Roles of the immune system in cancer: From tumor initiation to metastatic progression. Genes Dev. 2018, 32, 1267–1284. [Google Scholar] [CrossRef] [Green Version]
  20. You, H.; Baluszek, S.; Kaminska, B. Immune Microenvironment of Brain Metastases—Are Microglia and Other Brain Macrophages Little Helpers? Front. Immunol. 2019, 10, 1941. [Google Scholar] [CrossRef]
  21. Leibold, A.T.; Monaco, G.N.; Dey, M. The role of the immune system in brain metastasis. Curr. Neurobiol. 2019, 10, 33–48. [Google Scholar] [PubMed]
  22. Di Lorenzo, R.; Ahluwalia, M.S. Targeted therapy of brain metastases: Latest evidence and clinical implications. Ther. Adv. Med. Oncol. 2017, 9, 781–796. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Franchino, F.; Rudà, R.; Soffietti, R. Mechanisms and Therapy for Cancer Metastasis to the Brain. Front. Oncol. 2018, 8, 161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Pardridge, W.M. Blood–brain barrier delivery. Drug Discov. Today 2007, 12, 54–61. [Google Scholar] [CrossRef] [PubMed]
  25. Kabraji, S.; Ni, J.; Lin, N.U.; Xie, S.; Winer, E.P.; Zhao, J.J. Drug Resistance in HER2-Positive Breast Cancer Brain Metastases: Blame the Barrier or the Brain? Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2018, 24, 1795–1804. [Google Scholar] [CrossRef] [Green Version]
  26. Dong, R.; Ji, J.; Liu, H.; He, X. The evolving role of trastuzumab emtansine (T-DM1) in HER2-positive breast cancer with brain metastases. Crit. Rev. Oncol. Hematol. 2019, 143, 20–26. [Google Scholar] [CrossRef]
  27. Nieder, C.; Grosu, A.L.; Gaspar, L.E. Stereotactic radiosurgery (SRS) for brain metastases: A systematic review. Radiat. Oncol. 2014, 9, 155. [Google Scholar] [CrossRef] [Green Version]
  28. Protopapa, M.; Kouloulias, V.; Nikoloudi, S.; Papadimitriou, C.; Gogalis, G.; Zygogianni, A. From Whole-Brain Radiotherapy to Immunotherapy: A Multidisciplinary Approach for Patients with Brain Metastases from NSCLC. J. Oncol. 2019, 2019, 12. [Google Scholar] [CrossRef]
  29. Ulahannan, D.; Khalifa, J.; Faivre-Finn, C.; Lee, S.M. Emerging treatment paradigms for brain metastasis in non-small-cell lung cancer: An overview of the current landscape and challenges ahead. Ann. Oncol. 2017, 28, 2923–2931. [Google Scholar] [CrossRef]
  30. Hardesty, D.A.; Nakaji, P. The Current and Future Treatment of Brain Metastases. Front. Surg. 2016, 3, 30. [Google Scholar] [CrossRef] [Green Version]
  31. Papademetriou, I.T.; Porter, T. Promising approaches to circumvent the blood–brain barrier: Progress, pitfalls and clinical prospects in brain cancer. Ther. Deliv. 2015, 6, 989–1016. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Teleanu, D.M.; Chircov, C.; Grumezescu, A.M.; Volceanov, A.; Teleanu, R.I. Blood–brain Delivery Methods Using Nanotechnology. Pharmaceutics 2018, 10, 269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Dong, X. Current Strategies for Brain Drug Delivery. Theranostics 2018, 8, 1481–1493. [Google Scholar] [CrossRef] [PubMed]
  34. Gabathuler, R. Approaches to transport therapeutic drugs across the blood–brain barrier to treat brain diseases. Neurobiol. Dis. 2010, 37, 48–57. [Google Scholar] [CrossRef]
  35. Bellettato, C.M.; Scarpa, M. Possible strategies to cross the blood–brain barrier. Ital. J. Pediatrics 2018, 44, 131. [Google Scholar] [CrossRef]
  36. Pandey, P.K.; Sharma, A.K.; Gupta, U. Blood brain barrier: An overview on strategies in drug delivery, realistic in vitro modeling and in vivo live tracking. Tissue Barriers 2015, 4, e1129476. [Google Scholar] [CrossRef] [Green Version]
  37. Fortin, D.; Gendron, C.; Boudrias, M.; Garant, M.-P. Enhanced chemotherapy delivery by intraarterial infusion and blood–brain barrier disruption in the treatment of cerebral metastasis. Cancer 2007, 109, 751–760. [Google Scholar] [CrossRef]
  38. Kinoshita, M.; McDannold, N.; Jolesz, F.A.; Hynynen, K. Targeted delivery of antibodies through the blood–brain barrier by MRI-guided focused ultrasound. Biochem. Biophys. Res. Commun. 2006, 340, 1085–1090. [Google Scholar] [CrossRef]
  39. Borlongan, C.V.; Emerich, D.F. Facilitation of drug entry into the CNS via transient permeation of blood brain barrier: Laboratory and preliminary clinical evidence from bradykinin receptor agonist, Cereport. Brain Res. Bull. 2003, 60, 297–306. [Google Scholar] [CrossRef]
  40. Lipinski, C.A.; Lombardo, F.; Dominy, B.W.; Feeney, P.J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings1PII of original article: S0169-409X(96)00423-1. The article was originally published in Advanced Drug Delivery Reviews 23 (1997) 3–25.1. Adv. Drug Deliv. Rev. 2001, 46, 3–26. [Google Scholar] [CrossRef]
  41. Tajes, M.; Ramos-Fernández, E.; Weng-Jiang, X.; Bosch-Morató, M.; Guivernau, B.; Eraso-Pichot, A.; Salvador, B.; Fernàndez-Busquets, X.; Roquer, J.; Muñoz, F.J. The blood–brain barrier: Structure, function and therapeutic approaches to cross it. Molec. Membr. Biol. 2014, 31, 152–167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Parodi, A.; Rudzińska, M.; Deviatkin, A.A.; Soond, S.M.; Baldin, A.V.; Zamyatnin, A.A., Jr. Established and Emerging Strategies for Drug Delivery Across the Blood–brain Barrier in Brain Cancer. Pharmaceutics 2019, 11, 245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Pulgar, V.M. Transcytosis to Cross the Blood Brain Barrier, New Advancements and Challenges. Front. Neurosci. 2019, 12, 1019. [Google Scholar] [CrossRef] [PubMed]
  44. Neves, V.; Aires-da-Silva, F.; Corte-Real, S.; Castanho, M.A.R.B. Antibody Approaches To Treat Brain Diseases. Trends Biotechnol. 2015, 34, 36–48. [Google Scholar] [CrossRef]
  45. Khan, N.U.; Miao, T.; Ju, X.; Guo, Q.; Han, L. 6—Carrier-Mediated Transportation through BBB. In Brain Targeted Drug Delivery System; Gao, H., Gao, X., Eds.; Academic Press: New York, NY, USA, 2019; pp. 129–158. [Google Scholar] [CrossRef]
  46. Pavan, B.; Dalpiaz, A.; Ciliberti, N.; Vertuani, S. Progress in Drug Delivery to the Central Nervous System by the Prodrug Approach. Molecules 2008, 13, 1035–1065. [Google Scholar] [CrossRef] [Green Version]
  47. Witt, K.A.; Gillespie, T.J.; Huber, J.D.; Egleton, R.D.; Davis, T.P. Peptide drug modifications to enhance bioavailability and blood–brain barrier permeability. Peptides 2001, 22, 2329–2343. [Google Scholar] [CrossRef]
  48. Pardridge, W.M. Drug transport across the blood–brain barrier. J. Cereb Blood Flow. Metab. 2012, 32, 1959–1972. [Google Scholar] [CrossRef]
  49. Coloma, M.J.; Lee, H.J.; Kurihara, A.; Landaw, E.M.; Boado, R.J.; Morrison, S.L.; Pardridge, W.M. Transport Across the Primate Blood–brain Barrier of a Genetically Engineered Chimeric Monoclonal Antibody to the Human Insulin Receptor. Pharm. Res. 2000, 17, 266–274. [Google Scholar] [CrossRef]
  50. Pardridge, W. Blood–brain barrier drug targeting: The future of brain drug development. Mol. Interv. 2003, 3, 90–105. [Google Scholar] [CrossRef]
  51. Zhang, Y.; Pardridge, W.M. Blood–brain barrier targeting of BDNF improves motor function in rats with middle cerebral artery occlusion. Brain Res. 2006, 1111, 227–229. [Google Scholar] [CrossRef]
  52. Bien-Ly, N.; Yu, Y.J.; Bumbaca, D.; Elstrott, J.; Boswell, C.A.; Zhang, Y.; Luk, W.; Lu, Y.; Dennis, M.S.; Weimer, R.M.; et al. Transferrin receptor (TfR) trafficking determines brain uptake of TfR antibody affinity variants. J. Exp. Med. 2014, 211, 233–244. [Google Scholar] [CrossRef] [Green Version]
  53. Yu, Y.J.; Zhang, Y.; Kenrick, M.; Hoyte, K.; Luk, W.; Lu, Y.; Atwal, J.; Elliott, J.M.; Prabhu, S.; Watts, R.J.; et al. Boosting Brain Uptake of a Therapeutic Antibody by Reducing Its Affinity for a Transcytosis Target. Sci Transl Med. 2011, 3, 84ra44. [Google Scholar] [CrossRef] [PubMed]
  54. Yu, Y.J.; Atwal, J.K.; Zhang, Y.; Tong, R.K.; Wildsmith, K.R.; Tan, C.; Bien-Ly, N.; Hersom, M.; Maloney, J.A.; Meilandt, W.J.; et al. Therapeutic bispecific antibodies cross the blood–brain barrier in nonhuman primates. Sci. Transl. Med. 2014, 6, 261ra154. [Google Scholar] [CrossRef] [PubMed]
  55. Boado, R.J.; Zhang, Y.; Zhang, Y.; Pardridge, W.M. Humanization of anti-human insulin receptor antibody for drug targeting across the human blood–brain barrier. Biotechnol. Bioeng. 2007, 96, 381–391. [Google Scholar] [CrossRef] [PubMed]
  56. Vieira, D.B.; Gamarra, L.F. Multifunctional Nanoparticles for Successful Targeted Drug Delivery across the Blood–brain Barrier, Molecular Insight of Drug Design. IntechOpen 2018. [Google Scholar] [CrossRef] [Green Version]
  57. Tang, W.; Fan, W.; Lau, J.; Deng, L.; Shen, Z.; Chen, X. Emerging blood–brain-barrier-crossing nanotechnology for brain cancer theranostics. Chem. Soc. Rev. 2019, 48, 2967–3014. [Google Scholar] [CrossRef]
  58. Saraiva, C.; Praça, C.; Ferreira, R.; Santos, T.; Ferreira, L.; Bernardino, L. Nanoparticle-mediated brain drug delivery: Overcoming blood–brain barrier to treat neurodegenerative diseases. J. Control. Release 2016, 235, 34–47. [Google Scholar] [CrossRef] [Green Version]
  59. Loureiro, J.A.; Gomes, B.; Coelho, M.A.N.; Carmo Pereira, M.d.; Rocha, S. Targeting nanoparticles across the blood–brain barrier with monoclonal antibodies. Nanomedicine 2014, 9, 709–722. [Google Scholar] [CrossRef]
  60. Furtado, D.; Björnmalm, M.; Ayton, S.; Bush, A.I.; Kempe, K.; Caruso, F. Overcoming the Blood–Brain Barrier: The Role of Nanomaterials in Treating Neurological Diseases. Adv. Mater. 2018, 30, 1801362. [Google Scholar] [CrossRef] [Green Version]
  61. Abdul Razzak, R.; Florence, G.J.; Gunn-Moore, F.J. Approaches to CNS Drug Delivery with a Focus on Transporter-Mediated Transcytosis. Int. J. Mol. Sci. 2019, 20, 3108. [Google Scholar] [CrossRef] [Green Version]
  62. Mäger, I.; Meyer, A.H.; Li, J.; Lenter, M.; Hildebrandt, T.; Leparc, G.; Wood, M.J.A. Targeting blood–brain-barrier transcytosis—Perspectives for drug delivery. Neuropharmacology 2017, 120, 4–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Bickel, U. Antibody delivery through the blood–brain barrier. Adv. Drug Deliv. Rev. 1995, 15, 53–72. [Google Scholar] [CrossRef]
  64. Kumagai, A.; Eisenberg, J.; Pardridge, W. Adsorptive-mediated transcytosis of cationized albumin and a beta-endorphin-cationized albumin chimeric peptide by isolated brain capillaries. Model system of blood–brain barrier transport. J. Biol. Chem. 1987, 262, 15414–15419. [Google Scholar]
  65. Hervé, F.; Ghinea, N.; Scherrmann, J.-M. CNS Delivery Via Adsorptive Transcytosis. AAPS J. 2008, 10, 455–472. [Google Scholar] [CrossRef] [Green Version]
  66. Oller-Salvia, B.; Sanchez-Navarro, M.; Giralt, E.; Teixido, M. Blood–brain barrier shuttle peptides: An emerging paradigm for brain delivery. Chem. Soc. Rev. 2016, 45, 4690–4707. [Google Scholar] [CrossRef] [Green Version]
  67. McCully, M.; Sanchez-Navarro, M.; Teixido, M.; Giralt, E. Peptide Mediated Brain Delivery of Nano- and Submicroparticles: A Synergistic Approach. Curr. Pharm. Des. 2018, 24, 1366–1376. [Google Scholar] [CrossRef]
  68. Lagassé, H.A.D.; Alexaki, A.; Simhadri, V.L.; Katagiri, N.H.; Jankowski, W.; Sauna, Z.E.; Kimchi-Sarfaty, C. Recent advances in (therapeutic protein) drug development. F1000Research 2017, 6, 113. [Google Scholar] [CrossRef] [Green Version]
  69. Lee, J.I.; Zhang, L.; Men, A.Y.; Kenna, L.A.; Huang, S.M. CYP-Mediated Therapeutic Protein-Drug Interactions. Clin. Pharm. 2010, 49, 295–310. [Google Scholar] [CrossRef]
  70. Cavaco, M.; Goncalves, J. Interactions between therapeutic proteins and small molecules: The shared role of perpetrators and victims. Clin. Pharm. Ther. 2016, 102, 649–661. [Google Scholar] [CrossRef]
  71. Moorthy, B.S.; Xie, B.; Moussa, E.M.; Iyer, L.K.; Chandrasekhar, S.; Panchal, J.P.; Topp, E.M. Structure of Monoclonal Antibodies. In Biobetters: Protein Engineering to Approach the Curative; Rosenberg, A., Demeule, B., Eds.; Springer: New York, NY, USA, 2015; pp. 81–89. [Google Scholar] [CrossRef]
  72. Roopenian, D.C.; Akilesh, S. FcRn: The neonatal Fc receptor comes of age. Nat. Rev. Immunol. 2007, 7, 715–725. [Google Scholar] [CrossRef]
  73. Weiner, L.M.; Surana, R.; Wang, S. Antibodies and cancer therapy: Versatile platforms for cancer immunotherapy. Nat. Rev. Immunol. 2010, 10, 317–327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Dobson, C.L.; Devine, P.W.A.; Phillips, J.J.; Higazi, D.R.; Lloyd, C.; Popovic, B.; Arnold, J.; Buchanan, A.; Lewis, A.; Goodman, J.; et al. Engineering the surface properties of a human monoclonal antibody prevents self-association and rapid clearance in vivo. Sci. Rep. 2016, 6, 38644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Rodgers, K.R.; Chou, R.C. Therapeutic monoclonal antibodies and derivatives: Historical perspectives and future directions. Biotechnol. Adv. 2016, 34, 1149–1158. [Google Scholar] [CrossRef] [PubMed]
  76. Shuptrine, C.; Surana, R.; Weiner, L.M. Monoclonal Antibodies for the Treatment of Cancer. Semin. Cancer Biol. 2012, 22, 3–13. [Google Scholar] [CrossRef] [Green Version]
  77. Stoermer, K.A.; Morrison, T.E. Complement and Viral Pathogenesis. Virology 2011, 411, 362–373. [Google Scholar] [CrossRef] [Green Version]
  78. Kubota, T.; Niwa, R.; Satoh, M.; Akinaga, S.; Shitara, K.; Hanai, N. Engineered therapeutic antibodies with improved effector functions. Cancer Sci. 2009, 100, 1566–1572. [Google Scholar] [CrossRef]
  79. Irani, V.; Guy, A.J.; Andrew, D.; Beeson, J.G.; Ramsland, P.A.; Richards, J.S. Molecular properties of human IgG subclasses and their implications for designing therapeutic monoclonal antibodies against infectious diseases. Mol. Immunol. 2015, 67, 171–182. [Google Scholar] [CrossRef]
  80. Chames, P.; Van Regenmortel, M.; Weiss, E.; Baty, D. Therapeutic antibodies: Successes, limitations and hopes for the future. Br. J. Pharm. 2009, 157, 220–233. [Google Scholar] [CrossRef]
  81. Xenaki, K.T.; Oliveira, S.; van Bergen en Henegouwen, P.M.P. Antibody or Antibody Fragments: Implications for Molecular Imaging and Targeted Therapy of Solid Tumors. Front. Immunol. 2017, 8, 1287. [Google Scholar] [CrossRef]
  82. Bates, A.; Power, C.A. David vs. Goliath: The Structure, Function, and Clinical Prospects of Antibody Fragments. Antibodies 2019, 8, 28. [Google Scholar] [CrossRef] [Green Version]
  83. Brandstadter, R.; Katz Sand, I. The use of natalizumab for multiple sclerosis. Neuropsychiatr. Dis. Treat. 2017, 13, 1691–1702. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Lipton, R.B.; Tepper, S.J.; Reuter, U.; Silberstein, S.; Stewart, W.F.; Nilsen, J.; Leonardi, D.K.; Desai, P.; Cheng, S.; Mikol, D.D.; et al. Erenumab in chronic migraine. Neurology 2019, 92, e2250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Stüve, O.; Bennett, J.L. Pharmacological Properties, Toxicology and Scientific Rationale for the use of Natalizumab (Tysabri®) in Inflammatory Diseases. CNS Drug Rev. 2007, 13, 79–95. [Google Scholar] [CrossRef] [PubMed]
  86. Yaldizli, Ö.; Putzki, N. Natalizumab in the Treatment of Multiple Sclerosis. Ther. Adv. Neurol. Disord. 2009, 2, 115–128. [Google Scholar] [CrossRef] [Green Version]
  87. Ohlsson, L.; Haanes, K.A.; Kronvall, E.; Xu, C.; Snellman, J.; Edvinsson, L. Erenumab (AMG 334), a monoclonal antagonist antibody against the canonical CGRP receptor, does not impair vasodilatory or contractile responses to other vasoactive agents in human isolated cranial arteries. Cephalalgia 2019, 39, 1745–1752. [Google Scholar] [CrossRef] [PubMed]
  88. Goadsby, P.J.; Reuter, U.; Hallström, Y.; Broessner, G.; Bonner, J.H.; Zhang, F.; Sapra, S.; Picard, H.; Mikol, D.D.; Lenz, R.A. A Controlled Trial of Erenumab for Episodic Migraine. N. Engl. J. Med. 2017, 377, 2123–2132. [Google Scholar] [CrossRef] [PubMed]
  89. Zhou, Q.-H.; Fu, A.; Boado, R.J.; Hui, E.K.-W.; Lu, J.Z.; Pardridge, W.M. Receptor-mediated abeta amyloid antibody targeting to Alzheimer’s disease mouse brain. Mol. Pharm. 2011, 8, 280–285. [Google Scholar] [CrossRef] [Green Version]
  90. Zuchero, Y.J.; Chen, X.; Bien-Ly, N.; Bumbaca, D.; Tong, R.K.; Gao, X.; Zhang, S.; Hoyte, K.; Luk, W.; Huntley, M.A.; et al. Discovery of Novel Blood–brain Barrier Targets to Enhance Brain Uptake of Therapeutic Antibodies. Neuron 2016, 89, 70–82. [Google Scholar] [CrossRef] [Green Version]
  91. Sehlin, D.; Syvänen, S.; Ballanger, B.; Barthel, H.; Bischof, G.N.; Boche, D.; Boecker, H.; Bohn, K.P.; Borghammer, P.; Cross, D.; et al. Engineered antibodies: New possibilities for brain PET? Eur. J. Nucl. Med. Mol. Imaging 2019. [Google Scholar] [CrossRef] [Green Version]
  92. Camidge, D.R.; Lee, E.Q.; Lin, N.U.; Margolin, K.; Ahluwalia, M.S.; Bendszus, M.; Chang, S.M.; Dancey, J.; de Vries, E.G.E.; Harris, G.J.; et al. Clinical trial design for systemic agents in patients with brain metastases from solid tumours: A guideline by the Response Assessment in Neuro-Oncology Brain Metastases working group. Lancet Oncol. 2018, 19, e20–e32. [Google Scholar] [CrossRef]
  93. Wang, A.; Komiya, T. Brain metastasis as exclusion criteria in clinical trials involving extensive-stage small cell lung cancer. J. Cancer Res. Clin. Oncol. 2019, 145, 3099–3144. [Google Scholar] [CrossRef] [PubMed]
  94. Baik, C.S.; Chamberlain, M.C.; Chow, L.Q. Targeted Therapy for Brain Metastases in EGFR-Mutated and ALK-Rearranged Non-Small-Cell Lung Cancer. J. Thorac. Oncol. 2015, 10, 1268–1278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Lazaro, T.; Brastianos, P.K. Immunotherapy and targeted therapy in brain metastases: Emerging options in precision medicine. CNS Oncol. 2017, 6, 139–151. [Google Scholar] [CrossRef] [PubMed]
  96. Puttemans, J.; Lahoutte, T.; D’Huyvetter, M.; Devoogdt, N. Beyond the Barrier: Targeted Radionuclide Therapy in Brain Tumors and Metastases. Pharmaceutics 2019, 11, 376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Zhang, X.; Hao, J. Development of anticancer agents targeting the Wnt/β-catenin signaling. Am. J. Cancer Res. 2015, 5, 2344–2360. [Google Scholar] [PubMed]
  98. Li, Z.; Kang, Y. Emerging therapeutic targets in metastatic progression: A focus on breast cancer. Pharmacol. Ther. 2016, 161, 79–96. [Google Scholar] [CrossRef] [Green Version]
  99. Stalmans, S.; Bracke, N.; Wynendaele, E.; Gevaert, B.; Peremans, K.; Burvenich, C.; Polis, I.; De Spiegeleer, B. Cell-Penetrating Peptides Selectively Cross the Blood–brain Barrier In Vivo. PLoS ONE 2015, 10, e0139652. [Google Scholar] [CrossRef] [Green Version]
  100. AntibodySociety. Therapeutic Monoclonal Antibodies Approved or in Review in the EU or the US. Available online: https://www.antibodysociety.org/resources/approved-antibodies/ (accessed on 5 December 2019).
  101. De Angelis, F.; Chataway, J. Novel Multiple Sclerosis Drugs in the Pipeline. Clin. Pharmacol. Ther. 2019, 105, 1082–1090. [Google Scholar] [CrossRef]
  102. Jovčevska, I.; Muyldermans, S. The Therapeutic Potential of Nanobodies. BioDrugs 2019. [Google Scholar] [CrossRef] [Green Version]
  103. Cummings, J.; Lee, G.; Ritter, A.; Sabbagh, M.; Zhong, K. Alzheimer’s disease drug development pipeline: 2019. Alzheimer’s Dement. (N.Y.) 2019, 5, 272–293. [Google Scholar] [CrossRef]
  104. Kaplon, H.; Reichert, J.M. Antibodies to watch in 2019. mAbs 2019, 11, 219–238. [Google Scholar] [CrossRef] [PubMed]
  105. Guo, Z.; Peng, H.; Kang, J.; Sun, D. Cell-penetrating peptides: Possible transduction mechanisms and therapeutic applications. Biomed. Rep. 2016, 4, 528–534. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Avci, F.G.; Akbulut, B.S.; Ozkirimli, E. Membrane Active Peptides and Their Biophysical Characterization. Biomolecules 2018, 8, 77. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Bolintineanu, D.S.; Kaznessis, Y.N. Computational studies of protegrin antimicrobial peptides: A review. Peptides 2011, 32, 188–201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Madani, F.; Lindberg, S.; Langel, U.; Futaki, S.; Gräslund, A. Mechanisms of cellular uptake of cell-penetrating peptides. J. Biophys. (Hindawi Publ. Corp. Online) 2011, 2011, 414729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Derakhshankhah, H.; Jafari, S. Cell penetrating peptides: A concise review with emphasis on biomedical applications. Biomed. Pharmacother. 2018, 108, 1090–1096. [Google Scholar] [CrossRef]
  110. Zhang, T.-T.; Li, W.; Meng, G.; Wang, P.; Liao, W. Strategies for transporting nanoparticles across the blood–brain barrier. J. Biomater. Sci. 2016, 4, 219–229. [Google Scholar] [CrossRef]
  111. Frankel, A.D.; Pabo, C.O. Cellular uptake of the tat protein from human immunodeficiency virus. Cell 1988, 55, 1189–1193. [Google Scholar] [CrossRef]
  112. Kristensen, M.; Brodin, B. Routes for drug translocation across the blood–brain barrier: Exploiting peptides as delivery vectors. J. Pharm. Sci. 2017, S0022-3549, 30361. [Google Scholar] [CrossRef] [Green Version]
  113. Duchardt, F.; Fotin-Mleczek, M.; Schwarz, H.; Fischer, R.; Brock, R. A Comprehensive Model for the Cellular Uptake of Cationic Cell-penetrating Peptides. Traffic 2007, 8, 848–866. [Google Scholar] [CrossRef]
  114. Schwarze, S.R.; Ho, A.; Vocero-Akbani, A.; Dowdy, S.F. In Vivo Protein Transduction: Delivery of a Biologically Active Protein into the Mouse. Science 1999, 285, 1569. [Google Scholar] [CrossRef]
  115. Antoniou, X.; Borsello, T. Cell Permeable Peptides: A Promising Tool to Deliver Neuroprotective Agents in the Brain. Pharmaceutics 2010, 3, 379–392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Zou, L.-L.; Ma, J.-L.; Wang, T.; Yang, T.-B.; Liu, C.-B. Cell-Penetrating Peptide-Mediated Therapeutic Molecule Delivery into the Central Nervous System. Curr. Neuropharmacol. 2013, 11, 197–208. [Google Scholar] [CrossRef]
  117. Ben-Zvi, T.; Yayon, A.; Gertler, A.; Monsonego-Ornan, E. Suppressors of cytokine signaling (SOCS) 1 and SOCS3 interact with and modulate fibroblast growth factor receptor signaling. J. Cell Sci. 2006, 119, 380. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Demeule, M.; Régina, A.; Ché, C.; Poirier, J.; Nguyen, T.; Gabathuler, R.; Castaigne, J.-P.; Béliveau, R. Identification and Design of Peptides as a New Drug Delivery System for the Brain. J. Pharm. Exp. 2008, 324, 1064. [Google Scholar] [CrossRef] [Green Version]
  119. Regina, A.; Demeule, M.; Tripathy, S.; Lachowicz, J. ANG4043, a Novel Brain-Penetrant Peptide-mAb Conjugate, Is Efficacious against HER2-Positive Intracranial Tumors in Mice. Mol. Cancer 2015, 14, 129–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Lim, S.; Kim, W.J.; Kim, Y.H.; Lee, S.; Koo, J.H.; Lee, J.A.; Yoon, H.; Kim, D.H.; Park, H.J.; Kim, H.M.; et al. dNP2 is a blood–brain barrier-permeable peptide enabling ctCTLA-4 protein delivery to ameliorate experimental autoimmune encephalomyelitis. Nat. Commun. 2015, 6, 8244. [Google Scholar] [CrossRef] [Green Version]
  121. Neves, V.; Aires-da-Silva, F.; Morais, M.; Gano, L.; Ribeiro, E.; Pinto, A.; Aguiar, S.; Gaspar, D.; Fernandes, C.; Correia, J.D.G.; et al. Novel peptides derived from Dengue virus capsid protein translocate reversibly the blood–brain barrier through a receptor-free mechanism. ACS Chem. Biol. 2017. [Google Scholar] [CrossRef]
  122. Côrte-Real, S.; Neves, V.; Oliveira, S.; Canhão, P.; Outeiro, T.; Castanho, M.; Aires da Silva, F. Antibody Molecules and Peptide Delivery Systems for Use in Alzheimer’S Disease and Related Disorders. WO2016120843A1, 29 January 2016. [Google Scholar]
  123. Cavaco, M.; Castanho Miguel, A.R.B.; Neves, V. Peptibodies: An elegant solution for a long-standing problem. Pept. Sci. 2018, 110, e23095. [Google Scholar] [CrossRef] [Green Version]
  124. Pyzik, M.; Rath, T.; Lencer, W.I.; Baker, K.; Blumberg, R.S. FcRn: The Architect Behind the Immune and Nonimmune Functions of IgG and Albumin. J. Immunol. 2015, 194, 4595. [Google Scholar] [CrossRef] [Green Version]
  125. Samec, N.; Jovcevska, I.; Stojan, J.; Zottel, A.; Liovic, M.; Myers, M.P.; Muyldermans, S.; Šribar, J.; Križaj, I.; Komel, R. Glioblastoma-specific anti-TUFM nanobody for in-vitro immunoimaging and cancer stem cell targeting. Oncotarget 2018, 9, 17282–17299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Hu, Y.; Liu, C.; Muyldermans, S. Nanobody-Based Delivery Systems for Diagnosis and Targeted Tumor Therapy. Front. Immunol. 2017, 8, 1442. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Steps in the formation of brain metastases (BM). Metastases formation begins in the microenvironment of the primary tumor with 1. metastatic clones developing, degrading the extracellular matrix (ECM), and suffering an epithelial–mesenchymal transition (EMT) to further detach from the connective tissue. 2. Subsequently, tumor cells invade and enter the circulation (intravation). 3. The dissemination within the vascular system drives tumor cells to distant sites, like the brain. 4. Then, they extravasate across the blood–brain barrier (BBB) and enter the brain parenchyma due to the release of proteolytic enzymes and cellular interactions. 5. Once inside the brain, cancer cells colonize the tissue and develop secondary tumors.
Figure 1. Steps in the formation of brain metastases (BM). Metastases formation begins in the microenvironment of the primary tumor with 1. metastatic clones developing, degrading the extracellular matrix (ECM), and suffering an epithelial–mesenchymal transition (EMT) to further detach from the connective tissue. 2. Subsequently, tumor cells invade and enter the circulation (intravation). 3. The dissemination within the vascular system drives tumor cells to distant sites, like the brain. 4. Then, they extravasate across the blood–brain barrier (BBB) and enter the brain parenchyma due to the release of proteolytic enzymes and cellular interactions. 5. Once inside the brain, cancer cells colonize the tissue and develop secondary tumors.
Pharmaceutics 12 00062 g001
Figure 2. Pathways across the blood–brain barrier (BBB). Representation of the BBB formed by the endothelial cells and their interaction with astrocytes. Different translocation routes are presented. (a) Tight junctions usually restrict the penetration of water-soluble compounds. (b) The large surface area of the lipid membranes of the endothelium offers an efficient diffuse route for lipid-soluble agents. (c) Several transport proteins (carriers) are present in the endothelium for glucose (Gluc-1), amino acids, nucleosides, and other substances. (d) Large molecules such as antibodies, lipoproteins, proteins, and peptides can only transverse the BBB by receptor-mediated transport (RMT). (e) The transport of native plasma proteins or peptides is limited, but cationization can increase their uptake by adsorptive-mediated transport (AMT).
Figure 2. Pathways across the blood–brain barrier (BBB). Representation of the BBB formed by the endothelial cells and their interaction with astrocytes. Different translocation routes are presented. (a) Tight junctions usually restrict the penetration of water-soluble compounds. (b) The large surface area of the lipid membranes of the endothelium offers an efficient diffuse route for lipid-soluble agents. (c) Several transport proteins (carriers) are present in the endothelium for glucose (Gluc-1), amino acids, nucleosides, and other substances. (d) Large molecules such as antibodies, lipoproteins, proteins, and peptides can only transverse the BBB by receptor-mediated transport (RMT). (e) The transport of native plasma proteins or peptides is limited, but cationization can increase their uptake by adsorptive-mediated transport (AMT).
Pharmaceutics 12 00062 g002
Figure 3. Main functions of therapeutic monoclonal antibodies (mAbs). mAbs have two antigen-binding fragments (Fabs) and one constant fragment crystallizable (Fc). The variable domain of the Fab confers specificity and binding affinity to either membrane receptors or soluble antigens. The Fc domain binds neonatal Fc receptor (FcRn), prolonging the half-life of mAbs; and connects immunoglobulin G (IgG) antibodies to immune effector mechanisms (antibody-dependent cell cytotoxicity—ADCC; and complement-dependent cytotoxicity—CDC) by engaging Fcγ receptors (FcγR) on immune cells, promoting cell lysis.
Figure 3. Main functions of therapeutic monoclonal antibodies (mAbs). mAbs have two antigen-binding fragments (Fabs) and one constant fragment crystallizable (Fc). The variable domain of the Fab confers specificity and binding affinity to either membrane receptors or soluble antigens. The Fc domain binds neonatal Fc receptor (FcRn), prolonging the half-life of mAbs; and connects immunoglobulin G (IgG) antibodies to immune effector mechanisms (antibody-dependent cell cytotoxicity—ADCC; and complement-dependent cytotoxicity—CDC) by engaging Fcγ receptors (FcγR) on immune cells, promoting cell lysis.
Pharmaceutics 12 00062 g003
Figure 4. Novel or innovative monoclonal antibody (mAb) strategies. Schematic representation of different antibody formats currently in research. (A) An intact IgG molecule alongside with various antibody fragments and their respective molecular weight. (B) Antibody-drug conjugates (ADC) are usually intact IgG molecules linked to a drug, toxin, or peptide to increase the cargo selectivity. (C) Fusion proteins are biopharmaceutical molecules where the binding domains can be derived from a receptor extracellular domain, cytokine, enzyme, and peptide. Depending on the IgG molecule, the Fc region is capable of FcγR and C1q binding, potentially enabling the fusion protein to initiate antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). (D) The scheme shows an intrabody made of a variable region of the light and heavy chain constituting the antigen-binding domain and an intracellular location domain to allow nuclear binding.
Figure 4. Novel or innovative monoclonal antibody (mAb) strategies. Schematic representation of different antibody formats currently in research. (A) An intact IgG molecule alongside with various antibody fragments and their respective molecular weight. (B) Antibody-drug conjugates (ADC) are usually intact IgG molecules linked to a drug, toxin, or peptide to increase the cargo selectivity. (C) Fusion proteins are biopharmaceutical molecules where the binding domains can be derived from a receptor extracellular domain, cytokine, enzyme, and peptide. Depending on the IgG molecule, the Fc region is capable of FcγR and C1q binding, potentially enabling the fusion protein to initiate antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). (D) The scheme shows an intrabody made of a variable region of the light and heavy chain constituting the antigen-binding domain and an intracellular location domain to allow nuclear binding.
Pharmaceutics 12 00062 g004
Table 1. Antibody therapeutics that are approved or in review for marketing for cancer or central-nervous-system (CNS) diseases.
Table 1. Antibody therapeutics that are approved or in review for marketing for cancer or central-nervous-system (CNS) diseases.
NameBrand NameTypeTargetFirst Indication Approved or ReviewedApproval Status
(EU|USA)
[fam-]trastuzumab deruxtecan-Humanized IgG1 ADCHER2HER2+ metastatic breast cancerN.A.|In review
Ado-trastuzumab emtansineKadcylaHumanized IgG1 ADCHER2Breast cancer2013|2012
AlemtuzumabLemtradaHumanized IgG1CD52Multiple sclerosis; chronic myeloid leukemia#2013; 2001 #|2014;2001 #
AtezolizumabTecentriqHumanized IgG1PD-L1Bladder cancer2017|2016
AvelumabBavencioHuman IgG1PD-L1Merkel cell carcinoma2017|2017
BevacizumabAvastinHumanized IgG1VEGFColorectal cancer2005|2004
BlinatumomabBlincytoMurine bispecific tandem scFvCD19, CD3Acute lymphoblastic leukemia2015|2014
Brentuximab vedotinAdcentrisChimeric IgG1 ADCCD30Hodgkin lymphoma, systemic anaplastic large cell lymphoma2012|2011
CemiplimabLibtayoHuman mAbPD-1Cutaneous squamous cell carcinoma2019|2018
CetuximabErbituxChimeric IgG1EGFRColorectal cancer2004|2004
DaclizumabZinbryta; ZenapaxHumanized IgG1IL-2RMultiple sclerosis #; prevention of kidney transplant rejection #2016 #; 1999 #|2016 #; 1997 #
DaratumumabDarzalexHuman IgG1CD38Multiple myeloma2016|2015
DinutuximabUnituxinChimeric IgG1GD2Neuroblastoma2015|2015
DurvalumabIMFINZIHuman IgG1PD-L1Bladder cancer2018|2017
EdrecolomabPanorexMurine IgG2aEpCAMColon cancer1995 *#|NA
ElotuzumabEmplicitiHumanized IgG1SLAMF7Multiple myeloma2016|2015
Enfortumab vedotin-Human IgG1 ADCNectin-4Urothelial cancerN.A.|In review
Eptinezumab-Humanized IgG1CGRPMigraine preventionN.A.|In review
ErenumabAimovigHuman IgG2CGRPMigraine prevention2018|2018
FremanezumabAjovyHumanized IgG2CGRPMigraine prevention2019|2018
GalcanezumabEmgalityHumanized IgG4CGRPMigraine prevention2018|2018
Gemtuzumab ozogamicinMylotargHumanized IgG4 ADCCD33Acute myeloid leukemia2018|2017; 2000 #
Ibritumomab tiuxetanZevalinMurine IgG1CD20Non-Hodgkin lymphoma2004|2002
IdarucizumabPraxbindHumanized FabDabigatranReversal of dabigatran-induced anticoagulation2015|2015
Inotuzumab ozogamicinBESPONSAHumanized IgG4 ADCCD22Acute lymphoblastic leukemia2017|2017
IpilimumabYervoyHuman IgG1CTLA-4Metastatic melanoma2011|2011
Isatuximab-Humanized IgG1CD38Multiple myelomaIn review|In review
NatalizumabTysabriHumanized IgG4α4 integrinMultiple sclerosis2006|2004
NebacumabCentoxinHuman IgMEndotoxinGran-negative sepsis1991 *#|N.A.
NecitumumabPortrazzaHuman IgG1EGFRNon-small cell lung cancer2015|2015
NivolumabOpdivoHuman IgG4PD1Melanoma, non-small cell lung cancer2015|2014
ObinutuzumabGazyvaHumanized IgG1 Glycoengineered CD20Chronic lymphocytic leukemia2014|2013
OcrelizumabOCREVUSHumanized IgG1CD20Multiple sclerosis2018|2017
OfatumumabArzerraHuman IgG1CD20Chronic lymphocytic leukemia2010|2009
OlaratumabLartruvoHuman IgG1PDGFRαSoft tissue sarcoma2016|2016
PanitumumabVectibixHuman IgG2EGFRColorectal cancer2007|2006
PembrolizumabKeytrudaHumanized IgG4PD1Melanoma2015|2014
PertuzumabPerjetaHumanized IgG1HER2Breast cancer2013|2012
Polatuzumab vedotinPolivyHumanized IgG1 ADCCD79bDiffuse large B-cell lymphomaIn review|2019
RamucirumabCyramzaHuman IgG1VEGFR2Gastric cancer2014|2014
RituximabMabTheraChimeric IgG1CD20Non-Hodgkin lymphoma1998|1997
Sacituzumab govitecan-Humanized IgG1TROP-2Triple-negative breast cancerN.A.|In review
Tafasitamab-Humanized IgG1CD19Diffuse large B-cell lymphomaN.A.|In review
Tositumomab-l131BexxarMurine IgG2aCD20Non-Hodgkin lymphomaN.A.|2003 #
TrastuzumabHeceptinHumanized IgG1HER2Breast cancer2000|1998
ADC, Antibody–drug conjugate; CGRP, Calcitonin gene-related peptide; CTLA-4, Cytotoxic T-lymphocyte-associated protein 4; EGFR, Epidermal growth factor receptor; EpCAM, Epithelial cellular adhesion molecule; Fab, Fragment antigen-binding; HER2, Human epidermal growth factor receptor-2; mAb, Monoclonal antibody; PD1, Programmed cell death protein-1; PDGFR, Platelet-derived growth factor receptor; PD-L1, Programmed death-ligand 1; scFv, Single-chain fragment variable; SLAMF7, Signaling lymphocytic activation molecule F7; TROP-2, Tumor-associated calcium signal transducer 2; VEGF, Vascular endothelial growth factor; N.A. Not approved; * Country-specific approval; # Withdrawn or marketing discontinued. Adapted from [100].
Table 2. Antibody therapeutics that are in investigation for cancer or central-nervous-system (CNS) diseases.
Table 2. Antibody therapeutics that are in investigation for cancer or central-nervous-system (CNS) diseases.
NameTypeTargetClinical IndicationsMost Advanced Phase
(vic-)trastuzumab duocarmazineHumanized IgG1 ADCHER2Breast cancerPhase 3
[125I]-mAb 425Human mAbEGFRGlioblastomaPhase 2
[131I]-BC-2 mAbHuman mAbTenascinGlioblastomaPhase 1/2
[131I]-chTNT-1/B MAbHuman mAbDNA-histone H1 complexGlioblastomaPhase 1/2
[131I]-SGMIB anti-HER2 VHH1Humanized VHHHER2Breast cancerPhase 1
[188Re]-labeled
Nimotuzumab
Humanized mAbEGFRGlioblastomaPhase 1
[211At]-labeled
81C6 mAb
Human mAbTenascinGlioblastomaPhase 1/2
131I-omburtamabMurine mAb, radio-labeledBT-H3Neuroblastoma central nervous system/leptomenigeal metastasesPhase 2/3
68-Ga-NOTA-anti-HER2 VHH1Humanized VHHHER2Brain metastases of breast carcinomaPhase 2
ABBV-8E12Human mAbTau proteinAlzheimer’s diseasePhase 2
AducanumabHuman IgG1Amyloid betaAlzheimer’s diseasePhase 3
AL002Human mAbTREM2 receptorAlzheimer’s diseasePhase 1
AL003Human mAbSIGLEC-3Alzheimer’s diseasePhase 1
ALX-0651Humanized VHHCXCR4-Phase 1
AndecaliximabHumanized IgG4MMP9Gastric or gastroesophageal junction adenocarcinomaPhase 3
BAT8001Humanized IgG1 ADCHER2Breast cancerPhase 3
BCD-100Human mAbPD-1MelanomaPhase 2/3
BernarituzumabHumanized IgG1FGFR2bGastric or gastroesophageal junction adenocarcinomaPhase 3
BIB092Human mAbTau proteinAlzheimer’s diseasePhase 2
biotin-coupled BC-4 +
Avidin + [90Y]-Biotin
Human mAbTenascinGlioblastomaPhase 1/2
Bispecific nb-derived CAR-T cellsBispecific Humanized tandem VHCD19/CD20Refractory/relapsed B-cell lymphomaPhase 1
CamrelizumabHumanized IgG4PD-1Hodgkin’s lymphoma, hepatocellular carcinomaPhase 3
CarotuximabChimeric IgG1EndoglinAngiosarcomaPhase 3
CrenezumabHumanized IgG4Amyloid betaAlzheimer’s diseasePhase 3
CS1001Human mAbPD-L1Non-small cell lung cancerPhase 3
Depatuxizumab mafodotinIgG1 ADCEGFRGlioblastomaPhase 2b/3
DonanemabHumanized IgG1 mAbAmyloid betaAlzheimer’s diseasePhase 2
Enfortumab vedotinHuman IgG1 ADCNectin-4Urothelial cancerPhase 3
EptinezumabHumanized IgG1CGRPEpisodic migrainesPhase 3
GantenerumabHuman IgG1Amyloid betaAlzheimer’s diseasePhase 3
I-131-BC8Murine IgG1, radio-labeledCD45Ablation of bone marrow to hematopoietic cell transplantation in AML patientsPhase 3
IBI308Human mAbPD-1Squamous cell non-small cell lung cancerPhase 3
IsatuximabHumanized IgG1CD38Multiple myelomaPhase 3
JNJ-63733657Human mAbTau proteinAlzheimer’s diseasePhase 1
KN035mAb single domainPD-L1Bile tract carcinomaPhase 3
L19IL2 + L19TNFscFv conjugatesFibronectin extra-domain BMelanomaPhase 3
Loncastuximab tesirineHumanized IgG1 ADCCD19Diffuse large B-cell lymphomaPhase 2
MargetuximabChimeric IgG1HER2Breast cancerPhase 3
Mirvetuximab soravtansineIgG1 ADCFolate receptor 1Ovarian cancerPhase 3
NaxitamabHumanized mAbGD2High-risk neuroblastoma and refractory osteomedullary diseasePhase 3
OpicinumabHuman mAbLINGO-1Multiple sclerosis, acute optic neuritisPhase 2
Oportuzumab monatoxHumanized scFv immunotoxinEpCAMBladder cancerPhase 3
Polatuzumab vedotinHumanized IgG1 ADCCD79bDiffuse large B-cell lymphomaPhase 3
RelatlimabHuman mAbLAG-3MelanomaPhase 2/3
Rovalpituzumab tesirineHumanized IgG1 ADCDLL3Small cell lung cancerPhase 3
SemorinemabHumanized IgG4Tau proteinAlzheimer’s diseasePhase 2
SolanezumabHumanized IgG1 mAbMonomersAlzheimer’s diseasePhase 3
SpartalizumabHumanized IgG4PD1MelanomaPhase 3
TisdelizumabHumanized mAbPD1Non-small cell lung cancer, Hodgkin’s lymphomaPhase 3
Trastuzumab deruxtecanHumanized ADCHER2Breast cancer, HER2+ gastric or gastroesophageal junction adenocarcinomaPhase 3
TremelimumabHuman IgG2CTLA4Non-small cell lung, head & neck, urothelial cancerPhase 3
TSR-042Humanized mAbPD1Ovarian cancerPhase 3
UblituximabChimeric IgG1CD20Chronic lymphocytic leukemiaPhase 3
UtomilumabHuman IgG2CD137Diffuse large B-cell lymphomaPhase 3
XMAB-5574Humanized IgG1CD19Diffuse large B-cell lymphomaPhase 2/3
ZagotenemabHuman mAbTau proteinAlzheimer’s diseasePhase 2
ZolbetuximabChimeric IgG1Claudin-18.2Gastric or gastroesophageal junction adenocarcinomaPhase 3
ADC, Antibody–drug conjugate; AML, Acute myeloid leukemia; CGRP, Calcitonin gene-related peptide; CTLA-4, Cytotoxic T-lymphocyte-associated protein 4; CXCR4, Chemokine receptor type 4; DLL3, Delta-like protein 3; EGFR, Epidermal growth factor receptor; EpCAM, Epithelial cellular adhesion molecule; HER2, Human epidermal growth factor receptor-2; LAG-3, Lymphocyte-activation gene 3; mAb, Monoclonal antibody; MMP-9, Matrix metalloproteinase-9; PD1, Programmed cell death protein-1; PD-L1, Programmed death-ligand 1; scFv, single-domain fragment variable; TREM2, Triggering receptor expressed on myeloid cells 2. Adapted from [96,101,102,103,104].

Share and Cite

MDPI and ACS Style

Cavaco, M.; Gaspar, D.; ARB Castanho, M.; Neves, V. Antibodies for the Treatment of Brain Metastases, a Dream or a Reality? Pharmaceutics 2020, 12, 62. https://doi.org/10.3390/pharmaceutics12010062

AMA Style

Cavaco M, Gaspar D, ARB Castanho M, Neves V. Antibodies for the Treatment of Brain Metastases, a Dream or a Reality? Pharmaceutics. 2020; 12(1):62. https://doi.org/10.3390/pharmaceutics12010062

Chicago/Turabian Style

Cavaco, Marco, Diana Gaspar, Miguel ARB Castanho, and Vera Neves. 2020. "Antibodies for the Treatment of Brain Metastases, a Dream or a Reality?" Pharmaceutics 12, no. 1: 62. https://doi.org/10.3390/pharmaceutics12010062

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop