Next Article in Journal
A Polytherapy Strategy Using Vincristine and ALK Inhibitors to Sensitise EML4-ALK-Positive NSCLC
Next Article in Special Issue
Treatment of Malignant Adnexal Tumors of the Skin: A 12-Year Perspective
Previous Article in Journal
The Use of ctDNA for BRAF Mutation Testing in Routine Clinical Practice in Patients with Advanced Melanoma
Previous Article in Special Issue
Recent Advances on Immunohistochemistry and Molecular Biology for the Diagnosis of Adnexal Sweat Gland Tumors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Antibody–Drug Conjugates as an Emerging Therapy in Oncodermatology

1
Team “Biologie des Infections à Polyomavirus”, UMR INRAE ISP 1282, Université de Tours, 31 Avenue Monge, 37200 Tours, France
2
Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Josef-Schneider-Straße 2, 97080 Würzburg, Germany
3
Department of Pathology, Université de Tours, CHU de Tours, Avenue de la République, 37170 Chambray-les-Tours, France
4
McSAF, 1 rue Claude Thion, 37200 Tours, France
5
Team Innovation Moléculaire et Thérapeutique (IMT), Groupe Innovation et Ciblage Cellulaire (GICC) EA7501, Université de Tours, 31 Avenue Monge, 37200 Tours, France
6
Department of Dermatology, Université de Tours, CHU de Tours, Avenue de la République, 37170 Chambray-les-Tours, France
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(3), 778; https://doi.org/10.3390/cancers14030778
Submission received: 4 January 2022 / Revised: 28 January 2022 / Accepted: 1 February 2022 / Published: 2 February 2022
(This article belongs to the Special Issue Rare Skin Cancers: Recent Advances in Classification and Management)

Abstract

:

Simple Summary

Currently, the therapeutic arsenal to fight cancers is extensive. Among these, antibody–drug conjugates (ADCs) consist in an antibody linked to a cytotoxic agent, allowing a specific delivery to tumor cells. ADCs are an emerging class of therapeutics, with twelve FDA- and EMA-approved drugs for hematological and solid cancers. In recent years, tremendous progress has been observed in therapeutic approaches for advanced skin cancer patients. ADCs appear as an emerging therapeutic option in oncodermatology. After providing an overview of ADC design and development, the goal of this article is to review the potential ADC indications in the field of oncodermatology.

Abstract

Antibody–drug conjugates (ADCs) are an emerging class of therapeutics, with twelve FDA- and EMA-approved drugs for hematological and solid cancers. Such drugs consist in a monoclonal antibody linked to a cytotoxic agent, allowing a specific cytotoxicity to tumor cells. In recent years, tremendous progress has been observed in therapeutic approaches for advanced skin cancer patients. In this regard, targeted therapies (e.g., kinase inhibitors) or immune checkpoint-blocking antibodies outperformed conventional chemotherapy, with proven benefit to survival. Nevertheless, primary and acquired resistances as well as adverse events remain limitations of these therapies. Therefore, ADCs appear as an emerging therapeutic option in oncodermatology. After providing an overview of ADC design and development, the goal of this article is to review the potential ADC indications in the field of oncodermatology.

1. Introduction

In 1909, the German chemist Paul Ehrlich [1] provided the first description of “targeted therapy”. He proposed the so called “magic bullet” concept, allowing the delivery of a therapeutic molecule to a specific target without affecting healthy tissues [2]. Currently, targeted therapies are used in daily oncology practice including oncodermatology. As such, tyrosine kinase inhibitors (TKIs) targeting BRAF and MEK proteins block the constitutive activation of the MAPK pathway in patients with BRAF-mutated melanoma [3], therefore reducing maintenance, development or dissemination of the cancer. Similarly, sonic hedgehog pathway inhibitors (sonidegib and vismodegib) act by inhibiting the smoothened protein involved in hedgehog signal transduction which plays a crucial role in basal cell carcinoma (BCC) development.
Among targeted therapies, antibody–drug conjugates (ADCs) combine a monoclonal antibody (mAb) with a highly cytotoxic molecule, allowing its specific delivery to tumor cells [4]. In recent years, optimization of ADC technologies in order to increase their therapeutic performances and overcome their limitations as well as evaluation of their effects in combination with currently approved drugs have significantly expanded their use in oncology [5,6]. Indeed, twelve ADCs have been approved by the FDA for treatment of hematologic malignancies ((A) in Table 1) and solid tumors ((B) in Table 1). Moreover, the number of ADCs in development is steadily increasing, with 195 ongoing clinical trials (https://www.beacon-intelligence.com/, accessed on 8 December 2021). Overall, ADCs are currently being applied in all oncology areas including skin cancers.
The aim of the present review is to highlight current developments of ADCs in oncodermatology and identify potential opportunities in this field.

2. ADCs in Oncology

2.1. Structure of ADCs

ADCs consist in a cytotoxic drug, i.e., a payload bioconjugated through a linker to mAb-targeting tumor cell antigens [7] (Figure 1 and Figure 2). The specific combination of these components determines both the therapeutic performance and safety of the ADC [4]. The following parts aim to provide a brief overview of the features of the different ADC components and how these can impact efficacy and safety.

2.2. Targets

Target identification is a crucial point in ADC development [8,9]. Tumor-specific biomarkers previously used for diagnosis, as well as proteins overexpressed due to gene amplification or proteins involved in tumor aggressiveness [10] might be considered as potential ADC targets. Most ADCs bind to proteins expressed on the cell surface while targeting of intracellular tumor-associated proteins can also be achieved by using T-cell-receptor-like antibodies recognizing peptides in the context of presentation by MHC-I complexes [11]. In both cases, i.e., extra- and intracellular proteins, the specificity of the tumor antigen, antigen expression levels and antigen/ADC internalization [12] determine the performance of an ADC target [9].
To provide optimal payload delivery, high and homogenous expression of targeted antigens on the tumor cell surface is required [13]. By contrast, absent or low expression in healthy tissues [14] is expected in order to limit toxicities on physiologic cells (i.e., in order to avoid on-target off-tumor cytotoxicity). Nevertheless, several clinically approved ADCs such as brentuximab vedotin, targeting CD30 [15], polatuzumab vedotin, targeting CD79 [16], and inotuzumab ozogamicin, targeting CD22 [17], actually engage with ligands expressed by immune cells but still harbor therapeutic efficiency and acceptable tolerance. Moreover, most ADCs require internalization of the targeted surface antigen through receptor-mediated endocytosis and lysosomal degradation of the linker in order to deliver the payload [18]. One evasion mechanism that limits ADC performance is the downregulation of the targeted antigen expression, which obviously affects ADC binding to the target cells [9].
While antigens present on the cell surface are the most frequent candidates for ADC targets [19], secreted proteins/soluble antigens might additionally offer new opportunities by being more accessible. As a consequence, linker and drug have to be designed to be active in this context [14]. Some ADCs targeting non-internalizing soluble antigens, such as Tenascin-C splice variants or fibronectin, with maytansinoid and auristatins as payloads, have already demonstrated antitumor activity [20,21]. Importantly, targeting soluble antigens in the tumor microenvironment (TME) might impact not only tumor cells but also tumor stroma, extracellular matrix and blood vessels. Moreover, an association between bevacizumab, an antivascular endothelial growth factor (VEGF) monoclonal antibody, and an ADC has provided improved outcomes compared to a combination of bevacizumab with standard chemotherapy, in a phase Ib clinical study including patients with platinum-resistant ovarian cancer [22].

2.3. Antibodies

mAbs used in ADC structures are mostly of the IgG1 or IgG4 subtypes, and are chimeric or humanized in order to decrease their immunogenicity [4]. Both parts of the mAbs, i.e., the fragment antigen-binding (Fab) and fragment crystallizable regions (Fc region), contribute to the performance of an ADC. While affinity of a mAb for its epitope is determined by the Fab portion [10], the Fc part is determinant for mAb stability and interactions with immune cells such as antibody-dependent cell-mediated cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC) and antibody-dependent cell-mediated phagocytosis (ADCP). To this end, the Fc part interacts with neonatal Fc receptor (FcRn) [23], a recycling receptor determining both IgG half-life and biodistribution [24], or complement (C1q) implied in CDC or Fc receptors on immune effector cells (FcγR), a family of receptors which may trigger an immune response upon Fc binding [25]. The ADC interactions with immune cells might either constitute an opportunity to induce an antitumor response, or represent a potential side effect, resulting in toxicity on immune cells [14]. Accordingly, to improve or avoid immune functions (ADCC, CDC, and ADCP), genetic engineering introducing mutations in the Fc domain of ADCs is used [14,26]. The Fc mutations aim to promote or impair interactions between ADCs and immune cells, Fc receptors or complement (C1q) in order to prevent or improve their activation, such as increased ADCC with ADC glyco-engineering (i.e., afucosylation) [27,28]. On the other hand, other Fc modifications have been proposed to increase ADC half-life via FcRn-mediated ADC recycling [9].
Additional factors related to the payload and linker affect ADC pharmacokinetics (PK) and pharmacodynamics (PD) [29]. Notably, ADC hydrophobicity is associated with shorter half-life in serum [30] and because most of the payloads are hydrophobic, attachment to a hydrophilic antibody remains a challenge [31]. To overcome hydrophobicity and ADC aggregation, the most common ADC modification consists in glycosylation, a naturally occurring post-translational modification influencing the solubility, antigenicity and stability of proteins [32]. PEGylation, the addition of polyethylene glycol (PEG) to the linker in order to mask payloads hydrophobicity and to improve the PK of ADCs (i.e., reduction in aggregation, improvement in solubility and half-life [31]), has also been proposed [32].

2.4. Payloads

While radionucleotides, toxins [33] and cytokines [34] have been suggested as ADC payloads [35,36,37], most preclinical and FDA/EMA-approved ADCs are bioconjugated with small cytotoxic molecules [38] with a broad range of structures and mechanisms of action [4]. Among these, duocarmycins and pyrrolobenzodiazepines (PBD) dimers induce DNA alkylation by linking alkyl residues to AT-rich regions or guanine, leading to cell apoptosis [38]. Cell death can also be induced by calicheamicins inducing DNA double-strand breaks [38]. Maytansine derivatives (including DM1 and DM4) [39] and auristatins (monomethyl auristatin E (MMAE), monomethyl auristatin F (MMAF)) [7] are poisons which inhibit microtubule polymerization by targeting tubulins, resulting in G2/M arrest and apoptosis. A third family of cytotoxic agents inhibit the topoisomerase enzymes, which control DNA structure changes. Topoisomerase inhibitors block the cell cycle ligation step, which generates DNA single- and double-strand breaks, leading to apoptotic cell death [40]. Trastuzumab deruxtecan and sacituzumab govitecan are both approved topoisomerase inhibitor ADCs [40]. Currently, auristatins, maytansine derivatives and PBD/PNU (anthracyclines family) represent 29%, 25% and 2% of the global active ADCs, respectively (https://www.beacon-intelligence.com/, accessed on 8 December 2021). Overall, MMAE remains the most frequent payload used for ADC development, involved in 19% of active ADCs for which the payload is disclosed (https://www.beacon-intelligence.com/, accessed on 8 December 2021).
Beyond their direct cytotoxic tumor-targeted activity, highly membrane-permeable payloads, such as MMAE or calicheamicin, are able to induce a “bystander effect” [41] due to drug release from the primary target cells into the TME. The bystander effect enhances ADC efficacy by targeting not only stromal cells, which do not express the targeted antigen, but also antigen-negative tumor cells in cases of intratumoral heterogeneity [12] (Figure 2). By contrast, MMAF, another auristatin derivate, has low cell permeability due to its negative charge. While such properties limit the off-target toxicity of MMAF ADCs, their bystander activity remains low [12]. Regarding this limit, MMAF ADCs require high tumor expression of target antigen [42]. Nevertheless, a MMAF-conjugated ADC, belantamab mafodotin, was approved in 2020 by the FDA and the EMA for relapsed or refractory multiple myeloma treatment [43,44].

2.5. Linker

The linker is the ADC portion that connects antibody and drug. Depending on the payload and antibody properties, either cleavable or non-cleavable linkers are used. Cleavable linkers are used in combination with payloads being only fully active after lysosomal degradation (e.g., MMAE or calicheamicin) [38]. To prevent the cleavage of the linker in blood circulation resulting in unspecific cytotoxicity, the ADC linker has to be stable until the ADC reaches its target cell. To this end, cleavable linkers are usually sensitive to conditions specifically found in lysosomes (e.g., acidic condition or lysosomic proteases (cathepsin B, glycosidase, phosphatase)) or high intracellular glutathione concentrations [38] and thereby release the payload only after endocytosis is achieved, leading to death of the target cell [38] (Figure 2).
Regarding non-cleavable linkers, the drug is active after internalization and enzymatic digestion of the antibody into the lysosome, allowing the release of an active metabolite, amino acid–linker–drug complex (e.g., trastuzumab emtansine) [7]. Non-cleavable linkers allow increased ADC stability, leading to reduced toxicity in non-target tissues [7]. Payloads such as MMAF or DM1 are classically used in this setting.

2.6. Linker–Antibody Conjugation

The drug to antibody ratio (DAR), defined as the number of drug molecules attached to one mAb, is another important determinant for ADC performance [38]. The optimal DAR depends on the nature of the payload [45]. The paradigm that a DAR value of 4 is optimal for pharmacokinetics has been challenged by the recent approval of DAR 8 antibodies, in particular in light of improved hydrophobicity-masking technologies [46,47]. The technology connecting the mAb to the linker is crucial to obtain a homogeneous and controlled DAR (i.e., position of drugs and loading) [48]. Chemical linkers involving functional groups (i.e., bioconjugation head) are used to bind to mAb amino acids. First-generation bioconjugation technologies involved either N-hydroxysuccinimide (NHS) ester bioconjugation heads binding to lysine residues, or hydrazones, maleimides and thioethers binding to cysteines [49]. However, those bioconjugation heads led to the generation of heterogeneous ADCs, presenting a high variability of the DAR in the range of 0–8, and to multiple different linker positions [38].
More recently, site-specific conjugation, i.e., engineered cysteine residues, unnatural amino acids, or enzymatic conjugation through glycosyltransferases have been applied to obtain more homogeneous ADCs [7]. Indeed, site-specific conjugation of antibodies has been shown to improve the therapeutic index because it improves ADC pharmacokinetics by reducing the hydrophobicity of the linker–payload as well as prevent the release of payload in blood [48]. Moreover, site-specific conjugation with rebridging of antibody disulfide bonds results in stable and homogeneous ADCs with a controlled number and position of payload, and higher stability characteristics [50].
Interestingly, Van Geel et al. has shown that it is possible to link a payload to the native N-glycan residing at asparagine 297 and demonstrated in vitro and in vivo efficacy [51].
To conclude, ongoing optimization of ADC technology is likely to expand their application in oncology in the upcoming years, as illustrated below with ADCs currently approved or investigated in the field of oncodermatology.

3. ADCs in Cutaneous T-Cell Lymphoma

Cutaneous T-cell lymphoma (CTCL) was the first skin cancer for which a therapeutic ADC was approved. CTCLs are a group of extra-nodal non-Hodgkin’s lymphomas with primary cutaneous infiltration of malignant monoclonal T lymphocytes. Mycosis fungoides (MF) is the most common form of CTCLs, characterized by slow progression from patches to infiltrating plaques and eventually to cutaneous and extracutaneous tumors. Among CTCLs, Sézary syndrome (SS) is characterized by erythroderma, lymphadenopathy and systemic dissemination of malignant Sézary T cells [52]. Although rare, SS displays an aggressive course, with a median survival of 1 to 5 years [53]. In the case of advanced MF and SS, the only potentially curative treatment is allogenic hematopoietic stem cell transplantation [54]. CD30, a proliferation-promoting member of the TNF receptor family [54], is expressed in 12 to 23% of SS and MF cases [55]. Since CD30 expression is restricted in healthy human tissues to activated T, B and NK cells, it was chosen as a potential ADC target for treating advanced CTCLs [10] (Figure 3A). Accordingly, brentuximab vedotin (adcetris®), consisting of an anti-CD30 MMAE-conjugated ADC, with a cathepsin B cleavable linker, was developed (Table 2). Clinical benefit of brentuximab vedotin was demonstrated in CTCLs (stage IV SS and MF) with clinical objective response rates (ORR) of 70% [56,57]. Moreover, brentuximab vedotin achieved more durable responses (56% ORR lasting 4 months) than conventional treatments with methotrexate or bexarotene (12% ORR lasting 4 months) (Table 3). Consequently, brentuximab vedotin was approved by the FDA and the EMA in 2017 to treat CD30+ CTCLs after at least one line of systemic therapy.
However, resistance to MMAE was observed upon brentuximab vedotin treatment [58]. To overcome this resistance mechanism, a DM1-conjugated anti-CD30 ADC is currently in development. The anti-CD30 antibody is conjugated to DM1, a tubulin inhibitor, through antibody lysines by NHS ester non-cleavable linker (Table 2). This ADC has shown similar antitumoral effects to brentuximab vedotin without acquisition of payload resistance mechanism; moreover, the payload-bioconjugation modifications may have safety advantages comparing to brentuximab vedotin, without retro-Michael instability of maleimide. This ADC is currently evaluated in an ongoing clinical trial of patients with recurrent or refractory CD30+ hematological malignancies (NCT03894150).
Another approach includes the combination of brentuximab vedotin with other therapeutics in order to improve efficacy and/or circumvent resistance. For instance, combination with the PD-1 blocking antibody nivolumab is currently being investigated in two phase I/II trials [59] (NCT02581631 and NCT01703949). Similarly, a phase I study is currently investigating the association between brentuximab vedotin and romidepsin, a histone deacetylase inhibitor which is FDA approved as a monotherapy for CTCL patients (NCT02616965).
Since patients treated with brentuximab vedotin do not achieve improvements in long-term outcomes, new CTCL targets were identified and new ADCs were developed. Inducible Co-Stimulator (ICOS) is a T-cell costimulatory receptor involved in the development of CTCLs. High ICOS expression in CTCLs suggested it as a suitable target to develop an anti-ICOS MMAE-conjugated ADC [60]. Interestingly, a comparison between brentuximab vedotin and an anti-ICOS MMAE-conjugated ADC in a preclinical model of a CTCL xenograft showed a longer overall survival of mice [60]. Recently, cell surface proteome analysis on CTCL cell lines identified cell surface heat shock protein 70 (scHSP70) as highly expressed compared with normal T cells [61]. Then, an anti-scHSP70 MMAE-conjugated ADC was developed, and in vitro comparison with brentuximab vedotin showed similar activity against MS and SS cell lines [61]. However, under hypoxic conditions, such as a tumor microenvironment, scHSP70 expression was higher and cells were obviously more sensitive to the ADC—this result suggests an advantage over brentuximab vedotin [61].

4. ADCs in Melanoma

While BRAF/MEK inhibitors or immune checkpoint inhibitors (ICI) have revolutionized the treatment of patients with metastatic melanoma, approximately 50% of patients still experience fatal outcomes. Although 70% of patients with metastatic melanoma respond upon treatment with a combination of BRAF and MEK inhibitors [62], acquired resistance is frequent, especially in those with high tumoral burden at baseline. Acquired resistance to MAPK pathway inhibition is frequently associated with reactivation of the MAPK pathway due to secondary genetic (NRAS mutation, BRAF amplification, and MEK mutations) or epigenetic changes (Akt amplification, loss of PTEN, amplification of HGF, RTK (receptor tyrosine kinase), PDGFRβ, and IGF 1R) [3]. Metastatic melanoma patients can be treated with ICI such as the anti-PD1 antibodies pembrolizumab or nivolumab as monotherapies [63] or nivolumab combined with ipilimumab, an antibody targeting CTLA-4 on T cells [63]. Overall, primary resistance occurs in 40 to 65% of metastatic melanoma cases either treated with targeted therapies, i.e., BRAF/MEK inhibitors or ICI [64], highlighting the need for new therapeutic options [65]. In this context, several ADC strategies targeting tumor cell membrane proteins, mostly tyrosine kinase receptors, or soluble proteins, have been developed in recent years (Table 4). ADCs developed against melanoma are presented in three parts depending on their target types (Figure 3B).

4.1. Membrane Protein as Targets

Glycoprotein-NMB (gpNMB) is a transmembrane glycoprotein involved in tumor growth, tumor invasion and metastasis. Moreover, gpNMB exerts a direct inhibitory effect on activated T cells, impairing antitumor immunity and allowing immune evasion [66]. Accordingly, high gpNMB tumor expression levels are associated with poor clinical outcome [66]. In melanoma, 87% of tumors demonstrate membrane expression of gpNMB [67,68]. By contrast, in normal cells, gpNMB is restricted to intracellular compartments, therefore suggesting gpNMB as a promising ADC target in melanoma [66]. Accordingly, glembatumumab vedotin, a gpNMB-targeted MMAE-conjugated ADC [69], resulted in a 39% ORR in a phase I/II study conducted among patients with advanced melanoma refractory to ICI and BRAF/MEK inhibition, with acceptable toxicity (NCT00412828) [70]. In line with the observation that gpNMB expression is induced by the inhibition of MAPK pathway [71], combination of glembatumumab vedotin with MAPK pathway inhibitors demonstrated a synergistic therapeutic effect in a melanoma mouse model [71].
High and homogeneous expression levels of HER3, a tyrosine kinase receptor, have been observed in 65% of cutaneous melanomas [72]. HER3 is a member of the HER/EGFR family, and heterodimerization of HER3 with other HER family members (e.g., EGFR or HER2) leads to the activation of the PI3K/AKT pathway [73]. Accordingly, HER3 expression promotes tumor growth, and its overexpression was associated with impaired survival in melanoma [8,73]. Moreover, BRAF/MEK inhibitors induce FOXD3 expression, a transcription factor triggering HER3 expression. Hence, upregulation of HER3 by FOXD3 can promote adaptive resistance to BRAF inhibitors [74]. As a consequence, co-targeting HER3 and BRAF/MEK appears as an interesting option to overcome therapeutic resistances in BRAF-mutated melanoma [74]. However, unconjugated anti-HER3 antibodies have only shown limited antitumor activity justifying the use of anti-HER3 ADCs [75]. Indeed, an anti-HER3 MMAF-conjugate exhibited complete and durable antitumor responses in xenograft mice models [76]. Another anti-HER3 topoisomerase I inhibitor-conjugated ADC [77] is evaluated as monotherapy in phase I/II clinical trials for melanoma (NCT02980341).
Finally, Chen and collaborators identified the melanosomal protein, PMEL17, as a highly expressed marker restricted to melanoma cells. Accordingly, they generated an anti-PMEL17 MMAE-conjugated ADC which was cytotoxic in vitro and in vivo in a preclinical model [78].

4.2. Tyrosine Kinase Receptor

Endothelin B receptor (ETBR) is a member of the G protein-coupled receptor superfamily mediating tissue differentiation, growth, and repair, through the MAPK pathway [79]. ETBR has been reported to be overexpressed in metastatic melanoma compared to normal melanocytes [80]. ETBR signaling has been implicated in malignant transformation of melanocytes, suggesting it as an oncogenic driver for melanoma development [81]. Importantly, ETBR blockade results in inhibition of melanoma growth in vitro and in vivo [81], rendering ETBR as a possible target for an ADC [82]. Accordingly, an anti-ETBR MMAE-conjugated ADC is currently evaluated in a phase I clinical trial in patients with metastatic or unresectable melanoma (n = 53) (NCT01522664) [83]. Upon treatment, 32% of patients had stable disease beyond 6 months, irrespective of BRAF status [83]. Interestingly, in a preclinical model, combination of a single dose of ADC and daily intake of MAPK pathway inhibitors increased expression of ETBR and enhanced the therapeutic response [84].
Anexelekto (AXL) is a tyrosine kinase receptor frequently expressed in melanoma [85]. Its tumoral expression has been associated with a more invasive phenotype [85]. Antitumor activity of an anti-AXL MMAE-conjugated ADC was observed in a preclinical model, and again combination with BRAF/MEK inhibitors improved antitumoral cytotoxicity [86]. Importantly, a BRAF/MEK inhibitor-resistant but high-AXL-expressing tumor sub-population was killed by the anti-AXL ADC, while BRAF/MEK inhibition was specifically cytotoxic for the low-AXL-expressing tumor cells [86]. Following this promising preclinical study, a phase I/II clinical trial is ongoing to evaluate this ADC (called enapotamab vedotin) in solid tumors including melanoma (NCT02988817). Moreover, AXL could also be an interesting target for squamous cell carcinoma [87].
c-KIT is a tyrosine kinase receptor engaged by a ligand called stem cell factor. c-KIT is involved in regulation of apoptosis, differentiation and proliferation [88]. In cancer, c-KIT mutation or gene amplification results in uncontrolled proliferation and resistance to apoptosis [88]. High c-KIT expression is observed in 64 to 88% of melanomas [88]. So far, c-KIT-targeted tyrosine kinase inhibitors only showed limited clinical benefit in Kit-mutated metastatic melanoma, due to the emergence of secondary mutations [89]. Preclinical studies revealed a potent antitumor activity of an anti-c-KIT DM1-conjugated ADC on several c-KIT-positive solid cancers including melanoma and leukemia [90]. Nevertheless, in a phase I clinical trial (NCT02221505), acute hypersensitivity reactions triggered by degranulation of c-KIT-expressing mast cells led to the termination of the trial [90].

4.3. Soluble Target for ADCs

Non-internalizing ADCs are also investigated for melanoma treatment using an ADC targeting the galectin-3-binding protein (Gal-3BP), a metastasis-associated secreted protein [91] which is abundant in the TME [92]. Following treatment with anti-Gal-3BP ADC, complete tumor regression was achieved in a xenograft mouse melanoma model [91].

5. ADCs in Skin Carcinomas

ADC strategies have also been developed for treating skin carcinomas which are prone to display an aggressive course, mostly squamous cell carcinoma (SCC) and Merkel cell carcinoma (MCC).

5.1. Squamous Cell Carcinoma

SCC is a common skin cancer frequently induced by cumulative exposure to ultraviolet radiation [93]. SCC prognosis is mostly favorable after complete tumor resection while poor outcome is observed at the metastatic stage, with no demonstrated benefit of conventional chemotherapies or EGFR inhibitors on patient survival [93]. Recently, one PD-1 inhibitor, cemiplimab, was shown to provide a 47% ORR in patients with advanced or metastatic cutaneous SCCs, with durable responses in 61% of them [94,95]. PD-1 inhibitors are currently recommended as first-line treatment of advanced SCCs which are not candidates for surgery or radiation therapy [96].
Several targets have been identified as candidates for ADC therapy in lung or cervical SCCs [97] (Figure 3C). Most of the studies evaluated ADCs in head and neck SCCs. Overexpression of tissue factor (TF)—a transmembrane protein activating pro-survival pathways [98]—has been associated with high metastatic potential and poor outcome in various solid tumors [99]. High TF expression levels are also associated with increased expression of VEGF, enhancing tumor angiogenesis [98]. Tisotumab vedotin, a MMAE-conjugated anti-TF ADC with a protease-cleavable linker ((A) in Table 5), demonstrated a 16% ORR (95% CI 10.2−22.5) in a phase I/II study with 147 patients with multiple solid tumors, including SCCs of the head and neck. The safety profile appeared manageable although 27% of patients had a treatment-emergent serious adverse event related to the drug, and one death from pneumonia possibly related to the treatment [100]. A phase II study is ongoing to determine the efficacy and safety of tisotumab vedotin after failure of first-line standard of care therapy, in solid tumors including head and neck SCCs (NCT03485209).
Leucine-rich repeat containing 15 (LRRC15) is a marker of cancer-associated fibroblasts [101]. TGF-β, an immunosuppressive cytokine frequently expressed by tumor cells, induces LRRC15 expression in activated fibroblasts in the TME [102,103]. Targeting cancer-associated fibroblasts through LRRC15 in order to reduce immunosuppressive properties of the TME could overcome therapeutic resistances [104]. In line with the frequent LRRC15 expression in SCC cancer-associated fibroblasts (81% of cases) [101], an anti-LRRC15 MMAE-conjugated ADC, ABBV-085, triggered complete response in SCC xenograft (PDX) models [101]. Moreover, combination of this drug with an anti-PD1 antibody (mouse IgG2a, 17D2 clone) has shown potent activity in tumor models [101]. This ADC is currently being investigated in a phase I clinical trial including head and neck SCCs (NCT02001623) (Table 5A). Of note, LRRC15 is also expressed by melanoma tumor cells, which could constitute an additional application field for this ADC [101].
The major issue of ADCs targeting a molecule expressed by healthy tissues is illustrated by the outcome of the program development of bivatuzumab mertansine (Table 5A). This anti-CD44v6 DM1-conjugated ADC, CD44v6 being an aggressive variant of CD44, was assessed in four clinical trials including one enrolling head and neck SCC patients. After occurrence of a case of fatal toxic epidermal necrolysis together with other skin-related adverse events, clinical development of the ADC was discontinued [105]. This severe adverse event probably occurred due to CD44v6 expression by both SCC and normal squamous epithelium [105].

5.2. Merkel Cell Carcinoma

Merkel cell carcinoma (MCC) is an aggressive skin cancer induced either by UV light or the Merkel cell polyomavirus [106,107]. Until 2017, only cytotoxic chemotherapies, mostly platin salts and etoposide, were available for patients with advanced disease. In recent years, avelumab, an anti PD-L1 antibody, was shown to provide a 33% ORR in patients with metastatic disease, after failure of a first-line chemotherapy—most of these responses being durable. Avelumab was approved by the FDA and the EMA for treating patients with advanced metastatic MCCs [108]. Pembrolizumab was assessed in the first-line setting and was approved by the FDA in 2018 for treating advanced MCCs [109,110]. Although responses to ICI may be long-lasting, more than 50% of patients do not respond or develop secondary resistance [108].
CD56 has recently been suggested as a suitable target for antibody-dependent cellular cytotoxicity in MCCs, as it is expressed on the tumor cell surface by the majority of MCCs [111] (Figure 3C). Accordingly, an anti-CD56 DM1-conjugated ADC, lorvotuzumab mertansine (LM, also known as IMGN901), has been developed by covalently coupling the DM1 to the humanized anti-CD56 mAb, lorvotuzumab (huN901) (Table 5B). In a phase I clinical trial, this ADC demonstrated acceptable safety and tolerability in different CD56-expressing tumors [112,113]. Moreover, signs of clinical activity were especially detected in the four MCC-tested patients with two objective responses [112]. The following phase I/II clinical trial investigated the combination of carboplatine/etoposide/LM versus carboplatine/etoposide alone in patients with small-cell lung cancer [114]. This trial was first modified by a reduction in the dose of carboplatine/etoposide and finally discontinued because of safety concerns (NCT01237678) [114].
Recently, a new CD56-targeting MMAE-conjugated ADC, Adcitmer®, using a new bioconjugation approach has shown the control of MCC tumor growth in a mouse preclinical model [115].
Delta-like protein 3 (DLL3) is an inhibitory ligand of NOTCH receptors and is involved in neurogenesis during early embryonic development. DLL3 is upregulated in neuroendocrine tumor and has a minimal expression in normal tissues [116]. DLL3 expression was found in 91% of MCC patients [117] and high expression of DLL3 was associated with virus-positive MCCs [118]. Rovalpituzumab tesirine, an anti-DLL3 PBD-conjugated ADC [118], showed modest antitumor activity in patients with small-cell lung carcinoma treated in third lines [119] (NCT02674568), but such anti-DLL3 ADCs could represent a potential strategy for treating viro-positive MCCs.

6. Strengths and Weaknesses of ADCs: Challenges and Perspectives

When compared to other cancer therapies, notably conventional cytotoxic treatment, the use of ADCs is a vectorized therapy with low off-target toxicity. Nevertheless, ADCs could also have some drawbacks, due to recognition of the target on non-tumor cells, leading to “on-target off-tumor cytotoxicity”, limiting the therapeutic window in which ADCs can be applied [14]. Moreover, unspecific linker cleavage can cause drug delivery without antibody recognition, resulting in systemic toxicity, which represents another important limitation of ADCs [120]. Improvements in the production process as well as modifications in ADC structures may counteract these constraints [14]. In order to obtain stable ADCs, engineering of the bioconjugation head might contribute to linker stability improvement [9]. As an example, maleimide residue either included in the linker or the bioconjugation head might react with free thiol of plasma, e.g., albumin, resulting in the so called “retro-Michael reaction”. Hence, this retro-Michael reaction induces an unspecific release of the linker and drug in the plasma, which not only impacts the efficacy of the ADCs but also leads to off-target toxicity [120].
Several resistance mechanisms to ADCs have been described such as target antigen downregulation or mutations, and impairment of lysosomal degradation pathways. Moreover, ADC payloads can be rejected from tumor cells though multidrug resistance transporter efflux [121]. For instance, loss of CD30 expression can be observed in cutaneous CD30-positive lymphoid neoplasm treated with brentuximab vedotin [58,122]. Trogocytosis, i.e., the extraction of the antibody–epitope complexes by monocytes may induce resistance to the ADC in CD30-positive lymphoma [122].
A wide number of ADC optimizations have been developed to improve ADC performance. A new class of therapy aiming to improve the tumor specificity of mAbs, named probodies, consists in the optimization of mAbs used for ADC technology [123]. These antibodies are modified in order to mask the Fab paratopes, thereby limiting their activity in healthy tissue. By contrast, in the TME, high protease activity removes the masking peptide, allowing paratope–epitope recognition. Enhancement of the therapeutic index by such probody-based therapy was demonstrated in an EGFR-overexpressing mouse model [123]. The use of probodies in ADCs to enhance tumor cell-specific delivery of drugs has recently been tested [124]. In this respect, an anti-CD166 probody–drug conjugate has shown on-target on-tumor specificity in a preclinical study on lung cancer [125].
ADC antitumoral effect could be optimized by the use of innovative payloads or a new bioconjugation strategy. As an example, trastuzumab emtansine is indicated as second-line treatment for HER2-positive metastatic breast cancer. Interestingly, a second anti-HER2 ADC, trastuzumab deruxtecan [126], with eight topoisomerase inhibitors conjugated to trastuzumab through a cleavable linker was recently approved by the FDA and the EMA—this ADC has shown higher efficacy than trastuzumab emtansine, demonstrating the involvement of a payload and bioconjugation strategy in ADC performance [126].
Among the challenges, ADC toxicity (i.e., thrombocytopenia, neutropenia or peripheral neuropathy) should be known and controlled, for example by dose modulation [127]. Moreover, unexpected combinatorial effects, in particular atypical ocular toxicities of bleomycin and brentuximab vedotin observed for Hodgkin’s lymphoma, should be considered for future combination strategies [127].

7. Conclusions

Several targets in skin cancers are currently being investigated as candidates for ADC therapy, which allows the targeted delivery of a drug. ADC is a growing class of therapeutics in oncology, including one ADC approved for cutaneous T-cell lymphoma. In preclinical and clinical trials, further ADCs are currently evaluated for the treatment of CTCLs, melanoma, SCCs or MCCs. Various combinations of ADCs with other therapeutics are investigated to overcome limitations of these drugs due to intrinsic or acquired resistance. Increased knowledge on critical ADC features, such as target, antibody, linker and drug choice, allows a steady improvement in ADC design and better management of side effects.

Author Contributions

Conceptualization, C.E., T.K. and M.S.; methodology, C.E, T.K. and M.S.; validation, C.E., A.D., C.M., T.K. and M.S.; writing—original draft preparation, C.E., T.K. and M.S.; writing—review and editing, C.E., D.S., R.H., S.G., A.D., C.M., P.B., M.-C.V.-M., A.T., T.K. and M.S.; supervision, TK. and M.S. All authors have read and agreed to the published version of the manuscript.

Funding

This work did not receive any funding.

Conflicts of Interest

A.D., C.M. and M.-C.V.-M. are employees of McSAF, who is the owner of the patent Adcitmer®. A.T., T.K. and M.S. filed this patent. T.K. is A.D.’s husband. The other authors declare no conflict of interest.

Abbreviations

ADCantibody–drug conjugate
CTCLcutaneous T-cell lymphoma
DARdrug to antibody ratio
ETBRendothelin B receptor
FcRnneonatal Fc receptor
FDAfood and drug administration
Gal-3BPgalectin-3-binding protein
gpNMBglycoprotein-NMB
ICIimmune checkpoint inhibitors
LRRC15leucine-rich repeat containing 15 mAb: monoclonal antibody
MCCMerkel cell carcinoma
MFmycosis fungoides
MMAEmonomethyl auristatin E
MMAFmonomethyl auristatin F
PBDpyrrolobenzodiazepines
PDX patient-derived xenograft
RTKreceptor tyrosine kinase
SCCsquamous cell carcinoma
SSSézary syndrome
TFtissue factor
TMEtumor microenvironment

References

  1. Schwartz, R.S. Paul Ehrlich’s Magic Bullets. N. Engl. J. Med. 2004, 350, 1079–1080. [Google Scholar] [CrossRef] [PubMed]
  2. Strebhardt, K.; Ullrich, A. Paul Ehrlich’s Magic Bullet Concept: 100 Years of Progress. Nat. Rev. Cancer 2008, 8, 473–480. [Google Scholar] [CrossRef] [PubMed]
  3. Kakadia, S.; Yarlagadda, N.; Awad, R.; Kundranda, M.; Niu, J.; Naraev, B.; Mina, L.; Dragovich, T.; Gimbel, M.; Mahmoud, F. Mechanisms of Resistance to BRAF and MEK Inhibitors and Clinical Update of US Food and Drug Administration-Approved Targeted Therapy in Advanced Melanoma. OncoTargets Ther. 2018, 11, 7095–7107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Chau, C.H.; Steeg, P.S.; Figg, W.D. Antibody-Drug Conjugates for Cancer. Lancet 2019, 394, 793–804. [Google Scholar] [CrossRef]
  5. Tong, J.T.W.; Harris, P.W.R.; Brimble, M.A.; Kavianinia, I. An Insight into FDA Approved Antibody-Drug Conjugates for Cancer Therapy. Molecules 2021, 26, 5847. [Google Scholar] [CrossRef] [PubMed]
  6. Joubert, N.; Beck, A.; Dumontet, C.; Denevault-Sabourin, C. Antibody–Drug Conjugates: The Last Decade. Pharmaceuticals 2020, 13, 245. [Google Scholar] [CrossRef] [PubMed]
  7. Beck, A.; Goetsch, L.; Dumontet, C.; Corvaïa, N. Strategies and Challenges for the next Generation of Antibody-Drug Conjugates. Nat. Rev. Drug Discov. 2017, 16, 315–337. [Google Scholar] [CrossRef] [PubMed]
  8. Boni, V.; Sharma, M.R.; Patnaik, A. The Resurgence of Antibody Drug Conjugates in Cancer Therapeutics: Novel Targets and Payloads. Am. Soc. Clin. Oncol. Educ. Book Am. Soc. Clin. Oncol. Annu. Meet. 2020, 40, e58–e74. [Google Scholar] [CrossRef]
  9. Amani, N.; Dorkoosh, F.A.; Mobedi, H. ADCs, as Novel Revolutionary Weapons for Providing a Step Forward in Targeted Therapy of Malignancies. Curr. Drug Deliv. 2020, 17, 23–51. [Google Scholar] [CrossRef]
  10. Teicher, B.A. Antibody-Drug Conjugate Targets. Curr. Cancer Drug Targets 2009, 9, 982–1004. [Google Scholar] [CrossRef]
  11. Lai, J.; Wang, Y.; Wu, S.-S.; Ding, D.; Sun, Z.-Y.; Zhang, Y.; Zhou, J.; Zhou, Z.; Xu, Y.-C.; Pan, L.-Q.; et al. Elimination of Melanoma by Sortase A-Generated TCR-like Antibody-Drug Conjugates (TL-ADCs) Targeting Intracellular Melanoma Antigen MART-1. Biomaterials 2018, 178, 158–169. [Google Scholar] [CrossRef]
  12. Staudacher, A.H.; Brown, M.P. Antibody Drug Conjugates and Bystander Killing: Is Antigen-Dependent Internalisation Required? Br. J. Cancer 2017, 117, 1736–1742. [Google Scholar] [CrossRef]
  13. Damelin, M.; Zhong, W.; Myers, J.; Sapra, P. Evolving Strategies for Target Selection for Antibody-Drug Conjugates. Pharm. Res. 2015, 32, 3494–3507. [Google Scholar] [CrossRef] [PubMed]
  14. Hoffmann, R.M.; Coumbe, B.G.T.; Josephs, D.H.; Mele, S.; Ilieva, K.M.; Cheung, A.; Tutt, A.N.; Spicer, J.F.; Thurston, D.E.; Crescioli, S.; et al. Antibody Structure and Engineering Considerations for the Design and Function of Antibody Drug Conjugates (ADCs). OncoImmunology 2018, 7, e1395127. [Google Scholar] [CrossRef] [PubMed]
  15. Vaklavas, C.; Forero-Torres, A. Safety and Efficacy of Brentuximab Vedotin in Patients with Hodgkin Lymphoma or Systemic Anaplastic Large Cell Lymphoma. Ther. Adv. Hematol. 2012, 3, 209–225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Sehn, L.H.; Herrera, A.F.; Flowers, C.R.; Kamdar, M.K.; McMillan, A.; Hertzberg, M.; Assouline, S.; Kim, T.M.; Kim, W.S.; Ozcan, M.; et al. Polatuzumab Vedotin in Relapsed or Refractory Diffuse Large B-Cell Lymphoma. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2020, 38, 155–165. [Google Scholar] [CrossRef]
  17. Fayad, L.; Offner, F.; Smith, M.R.; Verhoef, G.; Johnson, P.; Kaufman, J.L.; Rohatiner, A.; Advani, A.; Foran, J.; Hess, G.; et al. Safety and Clinical Activity of a Combination Therapy Comprising Two Antibody-Based Targeting Agents for the Treatment of Non-Hodgkin Lymphoma: Results of a Phase I/II Study Evaluating the Immunoconjugate Inotuzumab Ozogamicin With Rituximab. J. Clin. Oncol. 2013, 31, 573–583. [Google Scholar] [CrossRef]
  18. Sievers, E.L.; Senter, P.D. Antibody-Drug Conjugates in Cancer Therapy. Annu. Rev. Med. 2013, 64, 15–29. [Google Scholar] [CrossRef]
  19. Thomas, A.; Teicher, B.A.; Hassan, R. Antibody–Drug Conjugates for Cancer Therapy. Lancet Oncol. 2016, 17, e254–e262. [Google Scholar] [CrossRef]
  20. Perrino, E.; Steiner, M.; Krall, N.; Bernardes, G.J.L.; Pretto, F.; Casi, G.; Neri, D. Curative Properties of Noninternalizing Antibody–Drug Conjugates Based on Maytansinoids. Cancer Res. 2014, 74, 2569–2578. [Google Scholar] [CrossRef] [Green Version]
  21. Gébleux, R.; Stringhini, M.; Casanova, R.; Soltermann, A.; Neri, D. Non-Internalizing Antibody–Drug Conjugates Display Potent Anti-Cancer Activity upon Proteolytic Release of Monomethyl Auristatin E in the Subendothelial Extracellular Matrix. Int. J. Cancer 2017, 140, 1670–1679. [Google Scholar] [CrossRef] [Green Version]
  22. O’Malley, D.M.; Matulonis, U.A.; Birrer, M.J.; Castro, C.M.; Gilbert, L.; Vergote, I.; Martin, L.P.; Mantia-Smaldone, G.M.; Martin, A.G.; Bratos, R.; et al. Phase Ib Study of Mirvetuximab Soravtansine, a Folate Receptor Alpha (FRα)-Targeting Antibody-Drug Conjugate (ADC), in Combination with Bevacizumab in Patients with Platinum-Resistant Ovarian Cancer. Gynecol. Oncol. 2020, 157, 379–385. [Google Scholar] [CrossRef] [PubMed]
  23. Castaneda, D.C.; Brachet, G.; Goupille, C.; Ouldamer, L.; Gouilleux-Gruart, V. The Neonatal Fc Receptor in Cancer FcRn in Cancer. Cancer Med. 2020, 9, 4736–4742. [Google Scholar] [CrossRef] [PubMed]
  24. Chen, N.; Wang, W.; Fauty, S.; Fang, Y.; Hamuro, L.; Hussain, A.; Prueksaritanont, T. The Effect of the Neonatal Fc Receptor on Human IgG Biodistribution in Mice. mAbs 2014, 6, 502–508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Nimmerjahn, F.; Ravetch, J.V. Fcgamma Receptors as Regulators of Immune Responses. Nat. Rev. Immunol. 2008, 8, 34–47. [Google Scholar] [CrossRef]
  26. Aguiar, S.; Dias, J.; Manuel, A.M.; Russo, R.; Gois, P.M.P.; da Silva, F.A.; Goncalves, J. Chimeric Small Antibody Fragments as Strategy to Deliver Therapeutic Payloads. Adv. Protein Chem. Struct. Biol. 2018, 112, 143–182. [Google Scholar] [CrossRef]
  27. Carter, P.J. Potent Antibody Therapeutics by Design. Nat. Rev. Immunol. 2006, 6, 343–357. [Google Scholar] [CrossRef] [PubMed]
  28. Gauzy-Lazo, L.; Sassoon, I.; Brun, M.-P. Advances in Antibody-Drug Conjugate Design: Current Clinical Landscape and Future Innovations. SLAS Discov. Adv. Sci. Drug Discov. 2020, 25, 843–868. [Google Scholar] [CrossRef]
  29. Lucas, A.T.; Robinson, R.; Schorzman, A.N.; Piscitelli, J.A.; Razo, J.F.; Zamboni, W.C. Pharmacologic Considerations in the Disposition of Antibodies and Antibody-Drug Conjugates in Preclinical Models and in Patients. Antibodies Basel Switz. 2019, 8, 3. [Google Scholar] [CrossRef] [Green Version]
  30. Hamblett, K.J.; Senter, P.D.; Chace, D.F.; Sun, M.M.C.; Lenox, J.; Cerveny, C.G.; Kissler, K.M.; Bernhardt, S.X.; Kopcha, A.K.; Zabinski, R.F.; et al. Effects of Drug Loading on the Antitumor Activity of a Monoclonal Antibody Drug Conjugate. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2004, 10, 7063–7070. [Google Scholar] [CrossRef] [Green Version]
  31. Buecheler, J.W.; Winzer, M.; Weber, C.; Gieseler, H. Alteration of Physicochemical Properties for Antibody-Drug Conjugates and Their Impact on Stability. J. Pharm. Sci. 2020, 109, 161–168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Buettner, M.J.; Shah, S.R.; Saeui, C.T.; Ariss, R.; Yarema, K.J. Improving Immunotherapy Through Glycodesign. Front. Immunol. 2018, 9, 2485. [Google Scholar] [CrossRef] [PubMed]
  33. Schrama, D.; Reisfeld, R.A.; Becker, J.C. Antibody Targeted Drugs as Cancer Therapeutics. Nat. Rev. Drug Discov. 2006, 5, 147–159. [Google Scholar] [CrossRef] [PubMed]
  34. King, D.M.; Albertini, M.R.; Schalch, H.; Hank, J.A.; Gan, J.; Surfus, J.; Mahvi, D.; Schiller, J.H.; Warner, T.; Kim, K.; et al. Phase I Clinical Trial of the Immunocytokine EMD 273063 in Melanoma Patients. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2004, 22, 4463–4473. [Google Scholar] [CrossRef]
  35. Teicher, B.A.; Chari, R.V.J. Antibody Conjugate Therapeutics: Challenges and Potential. Clin. Cancer Res. 2011, 17, 6389–6397. [Google Scholar] [CrossRef] [Green Version]
  36. Wu, A.M.; Senter, P.D. Arming Antibodies: Prospects and Challenges for Immunoconjugates. Nat. Biotechnol. 2005, 23, 1137–1146. [Google Scholar] [CrossRef]
  37. Milenic, D.E.; Brady, E.D.; Brechbiel, M.W. Antibody-Targeted Radiation Cancer Therapy. Nat. Rev. Drug Discov. 2004, 3, 488–499. [Google Scholar] [CrossRef] [Green Version]
  38. Leung, D.; Wurst, J.M.; Liu, T.; Martinez, R.M.; Datta-Mannan, A.; Feng, Y. Antibody Conjugates-Recent Advances and Future Innovations. Antibodies 2020, 9, 2. [Google Scholar] [CrossRef] [Green Version]
  39. Yaghoubi, S.; Karimi, M.H.; Lotfinia, M.; Gharibi, T.; Mahi-Birjand, M.; Kavi, E.; Hosseini, F.; Sepehr, K.S.; Khatami, M.; Bagheri, N.; et al. Potential Drugs Used in the Antibody–Drug Conjugate (ADC) Architecture for Cancer Therapy. J. Cell. Physiol. 2020, 235, 31–64. [Google Scholar] [CrossRef]
  40. Thomas, A.; Pommier, Y. Targeting Topoisomerase I in the Era of Precision Medicine. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2019, 25, 6581–6589. [Google Scholar] [CrossRef]
  41. Abdollahpour-Alitappeh, M.; Lotfinia, M.; Gharibi, T.; Mardaneh, J.; Farhadihosseinabadi, B.; Larki, P.; Faghfourian, B.; Sepehr, K.S.; Abbaszadeh-Goudarzi, K.; Abbaszadeh-Goudarzi, G.; et al. Antibody–Drug Conjugates (ADCs) for Cancer Therapy: Strategies, Challenges, and Successes. J. Cell. Physiol. 2019, 234, 5628–5642. [Google Scholar] [CrossRef] [PubMed]
  42. Smith, L.M.; Nesterova, A.; Alley, S.C.; Torgov, M.Y.; Carter, P.J. Potent Cytotoxicity of an Auristatin-Containing Antibody-Drug Conjugate Targeting Melanoma Cells Expressing Melanotransferrin/P97. Mol. Cancer Ther. 2006, 5, 1474–1482. [Google Scholar] [CrossRef] [Green Version]
  43. Tai, Y.-T.; Mayes, P.A.; Acharya, C.; Zhong, M.Y.; Cea, M.; Cagnetta, A.; Craigen, J.; Yates, J.; Gliddon, L.; Fieles, W.; et al. Novel Anti-B-Cell Maturation Antigen Antibody-Drug Conjugate (GSK2857916) Selectively Induces Killing of Multiple Myeloma. Blood 2014, 123, 3128–3138. [Google Scholar] [CrossRef] [PubMed]
  44. Lonial, S.; Lee, H.C.; Badros, A.; Trudel, S.; Nooka, A.K.; Chari, A.; Abdallah, A.-O.; Callander, N.; Sborov, D.; Suvannasankha, A.; et al. Longer Term Outcomes with Single-Agent Belantamab Mafodotin in Patients with Relapsed or Refractory Multiple Myeloma: 13-Month Follow-up from the Pivotal DREAMM-2 Study. Cancer 2021, 127, 4198–4212. [Google Scholar] [CrossRef]
  45. Walsh, S.J.; Bargh, J.D.; Dannheim, F.M.; Hanby, A.R.; Seki, H.; Counsell, A.J.; Ou, X.; Fowler, E.; Ashman, N.; Takada, Y.; et al. Site-Selective Modification Strategies in Antibody–Drug Conjugates. Chem. Soc. Rev. 2021, 50, 1305–1353. [Google Scholar] [CrossRef]
  46. Burke, P.J.; Hamilton, J.Z.; Jeffrey, S.C.; Hunter, J.H.; Doronina, S.O.; Okeley, N.M.; Miyamoto, J.B.; Anderson, M.E.; Stone, I.J.; Ulrich, M.L.; et al. Optimization of a PEGylated Glucuronide-Monomethylauristatin E Linker for Antibody-Drug Conjugates. Mol. Cancer Ther. 2017, 16, 116–123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Viricel, W.; Fournet, G.; Beaumel, S.; Perrial, E.; Papot, S.; Dumontet, C.; Joseph, B. Monodisperse Polysarcosine-Based Highly-Loaded Antibody-Drug Conjugates. Chem. Sci. 2019, 10, 4048–4053. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Junutula, J.R.; Raab, H.; Clark, S.; Bhakta, S.; Leipold, D.D.; Weir, S.; Chen, Y.; Simpson, M.; Tsai, S.P.; Dennis, M.S.; et al. Site-Specific Conjugation of a Cytotoxic Drug to an Antibody Improves the Therapeutic Index. Nat. Biotechnol. 2008, 26, 925–932. [Google Scholar] [CrossRef]
  49. Bargh, J.D.; Isidro-Llobet, A.; Parker, J.S.; Spring, D.R. Cleavable Linkers in Antibody–Drug Conjugates. Chem. Soc. Rev. 2019, 48, 4361–4374. [Google Scholar] [CrossRef]
  50. Juen, L.; Baltus, C.B.; Gély, C.; Feuillâtre, O.; Desgranges, A.; Viaud-Massuard, M.-C.; Martin, C. Innovative Bioconjugation Technology for Antibody-Drug Conjugates: Proof of Concept in a CD30-Positive Lymphoma Mouse Model. Bioconjugate Chem. 2021, 32, 595–606. [Google Scholar] [CrossRef]
  51. Van Geel, R.; Wijdeven, M.A.; Heesbeen, R.; Verkade, J.M.M.; Wasiel, A.A.; van Berkel, S.S.; van Delft, F.L. Chemoenzymatic Conjugation of Toxic Payloads to the Globally Conserved N-Glycan of Native MAbs Provides Homogeneous and Highly Efficacious Antibody–Drug Conjugates. Bioconjug. Chem. 2015, 26, 2233–2242. [Google Scholar] [CrossRef] [PubMed]
  52. Willemze, R.; Cerroni, L.; Kempf, W.; Berti, E.; Facchetti, F.; Swerdlow, S.H.; Jaffe, E.S. The 2018 Update of the WHO-EORTC Classification for Primary Cutaneous Lymphomas. Blood 2019, 133, 1703–1714. [Google Scholar] [CrossRef] [PubMed]
  53. Scarisbrick, J.J.; Prince, H.M.; Vermeer, M.H.; Quaglino, P.; Horwitz, S.; Porcu, P.; Stadler, R.; Wood, G.S.; Beylot-Barry, M.; Pham-Ledard, A.; et al. Cutaneous Lymphoma International Consortium Study of Outcome in Advanced Stages of Mycosis Fungoides and Sézary Syndrome: Effect of Specific Prognostic Markers on Survival and Development of a Prognostic Model. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2015, 33, 3766–3773. [Google Scholar] [CrossRef]
  54. Welborn, M.; Duvic, M. Antibody-Based Therapies for Cutaneous T-Cell Lymphoma. Am. J. Clin. Dermatol. 2019, 20, 115–122. [Google Scholar] [CrossRef] [PubMed]
  55. Pierce, J.M.R.; Mehta, A. Diagnostic, Prognostic and Therapeutic Role of CD30 in Lymphoma. Expert Rev. Hematol. 2017, 10, 29–37. [Google Scholar] [CrossRef] [PubMed]
  56. Mehra, T.; Ikenberg, K.; Moos, R.M.; Benz, R.; Nair, G.; Schanz, U.; Haralambieva, E.; Hoetzenecker, W.; Dummer, R.; French, L.E.; et al. Brentuximab as a Treatment for CD30+ Mycosis Fungoides and Sézary Syndrome. JAMA Dermatol. 2015, 151, 73–77. [Google Scholar] [CrossRef] [Green Version]
  57. Kim, Y.H.; Tavallaee, M.; Sundram, U.; Salva, K.A.; Wood, G.S.; Li, S.; Rozati, S.; Nagpal, S.; Krathen, M.; Reddy, S.; et al. Phase II Investigator-Initiated Study of Brentuximab Vedotin in Mycosis Fungoides and Sézary Syndrome with Variable CD30 Expression Level: A Multi-Institution Collaborative Project. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2015, 33, 3750–3758. [Google Scholar] [CrossRef]
  58. Chen, R.; Hou, J.; Newman, E.; Kim, Y.; Donohue, C.; Liu, X.; Thomas, S.H.; Forman, S.J.; Kane, S.E. CD30 Downregulation, MMAE Resistance, and MDR1 Upregulation Are All Associated with Resistance to Brentuximab Vedotin. Mol. Cancer Ther. 2015, 14, 1376–1384. [Google Scholar] [CrossRef] [Green Version]
  59. Merli, M.; Ferrario, A.; Maffioli, M.; Olivares, C.; Stasia, A.; Arcaini, L.; Passamonti, F. New Uses for Brentuximab Vedotin and Novel Antibody Drug Conjugates in Lymphoma. Expert Rev. Hematol. 2016, 9, 767–780. [Google Scholar] [CrossRef]
  60. Amatore, F.; Ortonne, N.; Lopez, M.; Barré, M.; Orlanducci, F.; Castellano, R.; Ingen-Housz-Oro, S.; De Croos, A.; Gorvel, L.; Goubard, A.; et al. ICOS Is Widely Expressed in Cutaneous T-Cell Lymphoma and Its Targeting Promotes Potent Killing of Malignant Cells. Blood 2021, 138, 790. [Google Scholar] [CrossRef]
  61. Choudhary, R.K.; Jones, R.J.; Kuiatse, I.; Wang, H.; Vega, F.; Bouska, A.C.; Lone, W.G.; Iqbal, J.; Orlowski, R.Z. A Novel Antibody Drug Conjugate (ADC) Targeting Cell Surface Heat Shock Protein 70 (CsHSP70) Is Active Against Pre-Clinical Models of Peripheral T-Cell Lymphoma (PTCL). Blood 2021, 138, 870. [Google Scholar] [CrossRef]
  62. Seth, R.; Messersmith, H.; Kaur, V.; Kirkwood, J.M.; Kudchadkar, R.; McQuade, J.L.; Provenzano, A.; Swami, U.; Weber, J.; Alluri, K.C.; et al. Systemic Therapy for Melanoma: ASCO Guideline. J. Clin. Oncol. 2020, 38, 3947–3970. [Google Scholar] [CrossRef]
  63. Albittar, A.A.; Alhalabi, O.; Oliva, I.C.G. Immunotherapy for Melanoma. Adv. Exp. Med. Biol. 2020, 1244, 51–68. [Google Scholar] [CrossRef] [PubMed]
  64. Gide, T.N.; Wilmott, J.S.; Scolyer, R.A.; Long, G.V. Primary and Acquired Resistance to Immune Checkpoint Inhibitors in Metastatic Melanoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2018, 24, 1260–1270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Kuske, M.; Westphal, D.; Wehner, R.; Schmitz, M.; Beissert, S.; Praetorius, C.; Meier, F. Immunomodulatory Effects of BRAF and MEK Inhibitors: Implications for Melanoma Therapy. Pharmacol. Res. 2018, 136, 151–159. [Google Scholar] [CrossRef]
  66. Rose, A.A.N.; Biondini, M.; Curiel, R.; Siegel, P.M. Targeting GPNMB with Glembatumumab Vedotin: Current Developments and Future Opportunities for the Treatment of Cancer. Pharmacol. Ther. 2017, 179, 127–141. [Google Scholar] [CrossRef]
  67. Williams, M.D.; Esmaeli, B.; Soheili, A.; Simantov, R.; Gombos, D.S.; Bedikian, A.Y.; Hwu, P. GPNMB Expression in Uveal Melanoma: A Potential for Targeted Therapy. Melanoma Res. 2010, 20, 184–190. [Google Scholar] [CrossRef]
  68. Taya, M.; Hammes, S.R. Glycoprotein Non-Metastatic Melanoma Protein B (GPNMB) and Cancer: A Novel Potential Therapeutic Target. Steroids 2018, 133, 102–107. [Google Scholar] [CrossRef]
  69. Tse, K.F.; Jeffers, M.; Pollack, V.A.; McCabe, D.A.; Shadish, M.L.; Khramtsov, N.V.; Hackett, C.S.; Shenoy, S.G.; Kuang, B.; Boldog, F.L.; et al. CR011, a Fully Human Monoclonal Antibody-Auristatin E Conjugate, for the Treatment of Melanoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2006, 12, 1373–1382. [Google Scholar] [CrossRef] [Green Version]
  70. Ott, P.A.; Hamid, O.; Pavlick, A.C.; Kluger, H.; Kim, K.B.; Boasberg, P.D.; Simantov, R.; Crowley, E.; Green, J.A.; Hawthorne, T.; et al. Phase I/II Study of the Antibody-Drug Conjugate Glembatumumab Vedotin in Patients with Advanced Melanoma. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2014, 32, 3659–3666. [Google Scholar] [CrossRef] [Green Version]
  71. Rose, A.A.N.; Annis, M.G.; Frederick, D.T.; Biondini, M.; Dong, Z.; Kwong, L.; Chin, L.; Keler, T.; Hawthorne, T.; Watson, I.R.; et al. MAPK Pathway Inhibitors Sensitize BRAF-Mutant Melanoma to an Antibody-Drug Conjugate Targeting GPNMB. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2016, 22, 6088–6098. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Ocana, A.; Vera-Badillo, F.; Seruga, B.; Templeton, A.; Pandiella, A.; Amir, E. HER3 Overexpression and Survival in Solid Tumors: A Meta-Analysis. J. Natl. Cancer Inst. 2013, 105, 266–273. [Google Scholar] [CrossRef] [Green Version]
  73. 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] [PubMed] [Green Version]
  74. Abel, E.V.; Basile, K.J.; Kugel, C.H.; Witkiewicz, A.K.; Le, K.; Amaravadi, R.K.; Karakousis, G.C.; Xu, X.; Xu, W.; Schuchter, L.M.; et al. Melanoma Adapts to RAF/MEK Inhibitors through FOXD3-Mediated Upregulation of ERBB3. J. Clin. Investig. 2013, 123, 2155–2168. [Google Scholar] [CrossRef] [PubMed]
  75. Zhang, N.; Chang, Y.; Rios, A.; An, Z. HER3/ErbB3, an Emerging Cancer Therapeutic Target. Acta Biochim. Biophys. Sin. 2016, 48, 39–48. [Google Scholar] [CrossRef] [Green Version]
  76. Capone, E.; Lamolinara, A.; D’Agostino, D.; Rossi, C.; De Laurenzi, V.; Iezzi, M.; Iacobelli, S.; Sala, G. EV20-Mediated Delivery of Cytotoxic Auristatin MMAF Exhibits Potent Therapeutic Efficacy in Cutaneous Melanoma. J. Control. Release Off. J. Control. Release Soc. 2018, 277, 48–56. [Google Scholar] [CrossRef]
  77. Hashimoto, Y.; Koyama, K.; Kamai, Y.; Hirotani, K.; Ogitani, Y.; Zembutsu, A.; Abe, M.; Kaneda, Y.; Maeda, N.; Shiose, Y.; et al. A Novel HER3-Targeting Antibody–Drug Conjugate, U3-1402, Exhibits Potent Therapeutic Efficacy through the Delivery of Cytotoxic Payload by Efficient Internalization. Clin. Cancer Res. 2019, 25, 7151–7161. [Google Scholar] [CrossRef] [Green Version]
  78. Chen, Y.; Chalouni, C.; Tan, C.; Clark, R.; Venook, R.; Ohri, R.; Raab, H.; Firestein, R.; Mallet, W.; Polakis, P. The Melanosomal Protein PMEL17 as a Target for Antibody Drug Conjugate Therapy in Melanoma. J. Biol. Chem. 2012, 287, 24082–24091. [Google Scholar] [CrossRef] [Green Version]
  79. Nelson, J.; Bagnato, A.; Battistini, B.; Nisen, P. The Endothelin Axis: Emerging Role in Cancer. Nat. Rev. Cancer 2003, 3, 110–116. [Google Scholar] [CrossRef]
  80. Saldana-Caboverde, A.; Kos, L. Roles of Endothelin Signaling in Melanocyte Development and Melanoma. Pigment. Cell Melanoma Res. 2010, 23, 160–170. [Google Scholar] [CrossRef] [Green Version]
  81. Bagnato, A.; Rosanò, L.; Spinella, F.; Di Castro, V.; Tecce, R.; Natali, P.G. Endothelin B Receptor Blockade Inhibits Dynamics of Cell Interactions and Communications in Melanoma Cell Progression. Cancer Res. 2004, 64, 1436–1443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Asundi, J.; Reed, C.; Arca, J.; McCutcheon, K.; Ferrando, R.; Clark, S.; Luis, E.; Tien, J.; Firestein, R.; Polakis, P. An Antibody-Drug Conjugate Targeting the Endothelin B Receptor for the Treatment of Melanoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2011, 17, 965–975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Sandhu, S.; McNeil, C.M.; LoRusso, P.; Patel, M.R.; Kabbarah, O.; Li, C.; Sanabria, S.; Flanagan, W.M.; Yeh, R.-F.; Brunstein, F.; et al. Phase I Study of the Anti-Endothelin B Receptor Antibody-Drug Conjugate DEDN6526A in Patients with Metastatic or Unresectable Cutaneous, Mucosal, or Uveal Melanoma. Investig. New Drugs 2019, 38, 844–854. [Google Scholar] [CrossRef] [PubMed]
  84. Asundi, J.; Lacap, J.A.; Clark, S.; Nannini, M.; Roth, L.; Polakis, P. MAPK Pathway Inhibition Enhances the Efficacy of an Anti-Endothelin B Receptor Drug Conjugate by Inducing Target Expression in Melanoma. Mol. Cancer Ther. 2014, 13, 1599–1610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Byers, L.A.; Diao, L.; Wang, J.; Saintigny, P.; Girard, L.; Peyton, M.; Shen, L.; Fan, Y.; Giri, U.; Tumula, P.K.; et al. An Epithelial-Mesenchymal Transition Gene Signature Predicts Resistance to EGFR and PI3K Inhibitors and Identifies Axl as a Therapeutic Target for Overcoming EGFR Inhibitor Resistance. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2013, 19, 279–290. [Google Scholar] [CrossRef] [Green Version]
  86. Boshuizen, J.; Koopman, L.A.; Krijgsman, O.; Shahrabi, A.; van den Heuvel, E.G.-; Ligtenberg, M.A.; Vredevoogd, D.W.; Kemper, K.; Kuilman, T.; Song, J.-Y.; et al. Cooperative Targeting of Melanoma Heterogeneity with an AXL Antibody-Drug Conjugate and BRAF/MEK Inhibitors. Nat. Med. 2018, 24, 203–212. [Google Scholar] [CrossRef]
  87. Cichoń, M.A.; Szentpetery, Z.; Caley, M.P.; Papadakis, E.S.; Mackenzie, I.C.; Brennan, C.H.; O’Toole, E.A. The Receptor Tyrosine Kinase Axl Regulates Cell-Cell Adhesion and Stemness in Cutaneous Squamous Cell Carcinoma. Oncogene 2014, 33, 4185–4192. [Google Scholar] [CrossRef] [Green Version]
  88. Miettinen, M.; Lasota, J. KIT (CD117): A Review on Expression in Normal and Neoplastic Tissues, and Mutations and Their Clinicopathologic Correlation. Appl. Immunohistochem. Mol. Morphol. AIMM 2005, 13, 205–220. [Google Scholar] [CrossRef]
  89. Gramza, A.W.; Corless, C.L.; Heinrich, M.C. Resistance to Tyrosine Kinase Inhibitors in Gastrointestinal Stromal Tumors. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2009, 15, 7510–7518. [Google Scholar] [CrossRef] [Green Version]
  90. Abrams, T.; Connor, A.; Fanton, C.; Cohen, S.B.; Huber, T.; Miller, K.; Hong, E.E.; Niu, X.; Kline, J.; Ison-Dugenny, M.; et al. Preclinical Antitumor Activity of a Novel Anti-c-KIT Antibody-Drug Conjugate against Mutant and Wild-Type c-KIT-Positive Solid Tumors. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2018, 24, 4297–4308. [Google Scholar] [CrossRef] [Green Version]
  91. Giansanti, F.; Capone, E.; Ponziani, S.; Piccolo, E.; Gentile, R.; Lamolinara, A.; Di Campli, A.; Sallese, M.; Iacobelli, V.; Cimini, A.; et al. Secreted Gal-3BP Is a Novel Promising Target for Non-Internalizing Antibody–Drug Conjugates. J. Control. Release 2019, 294, 176–184. [Google Scholar] [CrossRef] [PubMed]
  92. Cesinaro, A.M.; Trentini, G.P.; Natoli, C.; Grassadonia, A.; Tinari, N.; Iacobelli, S. Expression of the 90K Tumor-Associated Protein in Benign and Malignant Melanocytic Lesions. J. Investig. Dermatol. 2002, 119, 187–190. [Google Scholar] [CrossRef] [Green Version]
  93. Motaparthi, K.; Kapil, J.P.; Velazquez, E.F. Cutaneous Squamous Cell Carcinoma: Review of the Eighth Edition of the American Joint Committee on Cancer Staging Guidelines, Prognostic Factors, and Histopathologic Variants. Adv. Anat. Pathol. 2017, 24, 171–194. [Google Scholar] [CrossRef] [PubMed]
  94. Markham, A.; Duggan, S. Cemiplimab: First Global Approval. Drugs 2018, 78, 1841–1846. [Google Scholar] [CrossRef] [PubMed]
  95. Ahmed, S.R.; Petersen, E.; Patel, R.; Migden, M.R. Cemiplimab-Rwlc as First and Only Treatment for Advanced Cutaneous Squamous Cell Carcinoma. Expert Rev. Clin. Pharmacol. 2019, 12, 947–951. [Google Scholar] [CrossRef]
  96. Stratigos, A.J.; Garbe, C.; Dessinioti, C.; Lebbe, C.; Bataille, V.; Bastholt, L.; Dreno, B.; Fargnoli, M.C.; Forsea, A.M.; Frenard, C.; et al. European Interdisciplinary Guideline on Invasive Squamous Cell Carcinoma of the Skin: Part 2. Treatment. Eur. J. Cancer 2020, 128, 83–102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Breij, E.C.W.; de Goeij, B.E.C.G.; Verploegen, S.; Schuurhuis, D.H.; Amirkhosravi, A.; Francis, J.; Miller, V.B.; Houtkamp, M.; Bleeker, W.K.; Satijn, D.; et al. An Antibody-Drug Conjugate That Targets Tissue Factor Exhibits Potent Therapeutic Activity against a Broad Range of Solid Tumors. Cancer Res. 2014, 74, 1214–1226. [Google Scholar] [CrossRef] [Green Version]
  98. Kasthuri, R.S.; Taubman, M.B.; Mackman, N. Role of Tissue Factor in Cancer. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2009, 27, 4834–4838. [Google Scholar] [CrossRef] [Green Version]
  99. Van den Berg, Y.W.; Osanto, S.; Reitsma, P.H.; Versteeg, H.H. The Relationship between Tissue Factor and Cancer Progression: Insights from Bench and Bedside. Blood 2012, 119, 924–932. [Google Scholar] [CrossRef] [Green Version]
  100. De Bono, J.S.; Concin, N.; Hong, D.S.; Thistlethwaite, F.C.; Machiels, J.-P.; Arkenau, H.-T.; Plummer, R.; Jones, R.H.; Nielsen, D.; Windfeld, K.; et al. Tisotumab Vedotin in Patients with Advanced or Metastatic Solid Tumours (InnovaTV 201): A First-in-Human, Multicentre, Phase 1-2 Trial. Lancet Oncol. 2019, 20, 383–393. [Google Scholar] [CrossRef]
  101. Purcell, J.W.; Tanlimco, S.G.; Hickson, J.; Fox, M.; Sho, M.; Durkin, L.; Uziel, T.; Powers, R.; Foster, K.; McGonigal, T.; et al. LRRC15 Is a Novel Mesenchymal Protein and Stromal Target for Antibody-Drug Conjugates. Cancer Res. 2018, 78, 4059–4072. [Google Scholar] [CrossRef] [Green Version]
  102. Bierie, B.; Moses, H.L. Tumour Microenvironment: TGFbeta: The Molecular Jekyll and Hyde of Cancer. Nat. Rev. Cancer 2006, 6, 506–520. [Google Scholar] [CrossRef] [PubMed]
  103. Pickup, M.; Novitskiy, S.; Moses, H.L. The Roles of TGFβ in the Tumour Microenvironment. Nat. Rev. Cancer 2013, 13, 788–799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Kalluri, R.; Zeisberg, M. Fibroblasts in Cancer. Nat. Rev. Cancer 2006, 6, 392–401. [Google Scholar] [CrossRef] [PubMed]
  105. Riechelmann, H.; Sauter, A.; Golze, W.; Hanft, G.; Schroen, C.; Hoermann, K.; Erhardt, T.; Gronau, S. Phase I Trial with the CD44v6-Targeting Immunoconjugate Bivatuzumab Mertansine in Head and Neck Squamous Cell Carcinoma. Oral Oncol. 2008, 44, 823–829. [Google Scholar] [CrossRef]
  106. Feng, H.; Shuda, M.; Chang, Y.; Moore, P.S. Clonal Integration of a Polyomavirus in Human Merkel Cell Carcinoma. Science 2008, 319, 1096–1100. [Google Scholar] [CrossRef] [Green Version]
  107. Becker, J.C.; Stang, A.; DeCaprio, J.A.; Cerroni, L.; Lebbé, C.; Veness, M.; Nghiem, P. Merkel Cell Carcinoma. Nat. Rev. Dis. Primers 2017, 3, 17077. [Google Scholar] [CrossRef]
  108. D’Angelo, S.P.; Bhatia, S.; Brohl, A.S.; Hamid, O.; Mehnert, J.M.; Terheyden, P.; Shih, K.C.; Brownell, I.; Lebbé, C.; Lewis, K.D.; et al. Avelumab in Patients with Previously Treated Metastatic Merkel Cell Carcinoma: Long-Term Data and Biomarker Analyses from the Single-Arm Phase 2 JAVELIN Merkel 200 Trial. J. Immunother. Cancer 2020, 8, e000674. [Google Scholar] [CrossRef]
  109. Nghiem, P.T.; Bhatia, S.; Lipson, E.J.; Kudchadkar, R.R.; Miller, N.J.; Annamalai, L.; Berry, S.; Chartash, E.K.; Daud, A.; Fling, S.P.; et al. PD-1 Blockade with Pembrolizumab in Advanced Merkel-Cell Carcinoma. N. Engl. J. Med. 2016, 374, 2542–2552. [Google Scholar] [CrossRef]
  110. Nghiem, P.; Bhatia, S.; Lipson, E.J.; Sharfman, W.H.; Kudchadkar, R.R.; Brohl, A.S.; Friedlander, P.A.; Daud, A.; Kluger, H.M.; Reddy, S.A.; et al. Durable Tumor Regression and Overall Survival in Patients with Advanced Merkel Cell Carcinoma Receiving Pembrolizumab as First-Line Therapy. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2019, 37, 693–702. [Google Scholar] [CrossRef]
  111. Ollier, J.; Kervarrec, T.; Samimi, M.; Benlalam, H.; Aumont, P.; Vivien, R.; Touzé, A.; Labarrière, N.; Vié, H.; Clémenceau, B. Merkel Cell Carcinoma and Cellular Cytotoxicity: Sensitivity to Cellular Lysis and Screening for Potential Target Antigens Suitable for Antibody-Dependent Cellular Cytotoxicity. Cancer Immunol. Immunother. 2018, 67, 1209–1219. [Google Scholar] [CrossRef] [PubMed]
  112. Shah, M.H.; Lorigan, P.; O’Brien, M.E.R.; Fossella, F.V.; Moore, K.N.; Bhatia, S.; Kirby, M.; Woll, P.J. Phase I Study of IMGN901, a CD56-Targeting Antibody-Drug Conjugate, in Patients with CD56-Positive Solid Tumors. Investig. New Drugs 2016, 34, 290–299. [Google Scholar] [CrossRef]
  113. Ailawadhi, S.; Kelly, K.R.; Vescio, R.A.; Jagannath, S.; Wolf, J.; Gharibo, M.; Sher, T.; Bojanini, L.; Kirby, M.; Chanan-Khan, A. A Phase I Study to Assess the Safety and Pharmacokinetics of Single-Agent Lorvotuzumab Mertansine (IMGN901) in Patients with Relapsed and/or Refractory CD-56-Positive Multiple Myeloma. Clin. Lymphoma Myeloma Leuk. 2019, 19, 29–34. [Google Scholar] [CrossRef] [PubMed]
  114. Socinski, M.A.; Kaye, F.J.; Spigel, D.R.; Kudrik, F.J.; Ponce, S.; Ellis, P.M.; Majem, M.; Lorigan, P.; Gandhi, L.; Gutierrez, M.E.; et al. Phase 1/2 Study of the CD56-Targeting Antibody-Drug Conjugate Lorvotuzumab Mertansine (IMGN901) in Combination With Carboplatin/Etoposide in Small-Cell Lung Cancer Patients With Extensive-Stage Disease. Clin. Lung Cancer 2017, 18, 68–76.e2. [Google Scholar] [CrossRef]
  115. Esnault, C.; Leblond, V.; Martin, C.; Desgranges, A.; Baltus, C.B.; Aubrey, N.; Lakhrif, Z.; Lajoie, L.; Lantier, L.; Clémenceau, B.; et al. Adcitmer®, a New CD56-Targeting Monomethyl Auristatin E-Conjugated Antibody, Is a Potential Therapeutic Approach in Merkel Cell Carcinoma. Br. J. Dermatol. 2021. [Google Scholar] [CrossRef]
  116. Lashari, B.H.; Vallatharasu, Y.; Kolandra, L.; Hamid, M.; Uprety, D. Rovalpituzumab Tesirine: A Novel DLL3-Targeting Antibody-Drug Conjugate. Drugs RD 2018, 18, 255–258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Rand, J.; Balzer, B.L.; Frishberg, D.P.; Essner, R.; Shon, W. Prevalence of Delta-Like Protein 3 Expression in Merkel Cell Carcinoma. J. Am. Acad. Dermatol. 2019, 85, 749–750. [Google Scholar] [CrossRef]
  118. Xie, H.; Kaye, F.J.; Isse, K.; Sun, Y.; Ramoth, J.; French, D.M.; Flotte, T.J.; Luo, Y.; Saunders, L.R.; Mansfield, A.S. Delta-like Protein 3 Expression and Targeting in Merkel Cell Carcinoma. Oncol. 2020, 25, 810–817. [Google Scholar] [CrossRef]
  119. Morgensztern, D.; Besse, B.; Greillier, L.; Santana-Davila, R.; Ready, N.; Hann, C.L.; Glisson, B.S.; Farago, A.F.; Dowlati, A.; Rudin, C.M.; et al. Efficacy and Safety of Rovalpituzumab Tesirine in Third-Line and Beyond Patients with DLL3-Expressing, Relapsed/Refractory Small-Cell Lung Cancer: Results From the Phase II TRINITY Study. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2019, 25, 6958–6966. [Google Scholar] [CrossRef] [Green Version]
  120. Szijj, P.A.; Bahou, C.; Chudasama, V. Minireview: Addressing the Retro-Michael Instability of Maleimide Bioconjugates. Drug Discov. Today Technol. 2018, 30, 27–34. [Google Scholar] [CrossRef] [Green Version]
  121. Parslow, A.C.; Parakh, S.; Lee, F.-T.; Gan, H.K.; Scott, A.M. Antibody-Drug Conjugates for Cancer Therapy. Biomedicines 2016, 4, 14. [Google Scholar] [CrossRef] [PubMed]
  122. Goyal, A.; Patel, S.; Goyal, K.; Morgan, E.A.; Foreman, R.K. Variable Loss of CD30 Expression by Immunohistochemistry in Recurrent Cutaneous CD30+ Lymphoid Neoplasms Treated with Brentuximab Vedotin. J. Cutan. Pathol. 2019, 46, 823–829. [Google Scholar] [CrossRef] [PubMed]
  123. Desnoyers, L.R.; Vasiljeva, O.; Richardson, J.H.; Yang, A.; Menendez, E.E.M.; Liang, T.W.; Wong, C.; Bessette, P.H.; Kamath, K.; Moore, S.J.; et al. Tumor-Specific Activation of an EGFR-Targeting Probody Enhances Therapeutic Index. Sci. Transl. Med. 2013, 5, 207ra144. [Google Scholar] [CrossRef] [PubMed]
  124. Autio, K.A.; Boni, V.; Humphrey, R.W.; Naing, A. Probody Therapeutics: An Emerging Class of Therapies Designed to Enhance On-Target Effects with Reduced Off-Tumor Toxicity for Use in Immuno-Oncology. Clin. Cancer Res. 2020, 26, 984–989. [Google Scholar] [CrossRef] [Green Version]
  125. Chomet, M.; Schreurs, M.; Nguyen, M.; Howng, B.; Villanueva, R.; Krimm, M.; Vasiljeva, O.; van Dongen, G.A.M.S.; Vugts, D.J. The Tumor Targeting Performance of Anti-CD166 Probody Drug Conjugate CX-2009 and Its Parental Derivatives as Monitored by 89Zr-Immuno-PET in Xenograft Bearing Mice. Theranostics 2020, 10, 5815–5828. [Google Scholar] [CrossRef]
  126. Modi, S.; Saura, C.; Yamashita, T.; Park, Y.H.; Kim, S.-B.; Tamura, K.; Andre, F.; Iwata, H.; Ito, Y.; Tsurutani, J.; et al. Trastuzumab Deruxtecan in Previously Treated HER2-Positive Breast Cancer. N. Engl. J. Med. 2020, 382, 610–621. [Google Scholar] [CrossRef] [PubMed]
  127. Donaghy, H. Effects of Antibody, Drug and Linker on the Preclinical and Clinical Toxicities of Antibody-Drug Conjugates. mAbs 2016, 8, 659–671. [Google Scholar] [CrossRef]
Figure 1. Design of an antibody–drug conjugate and recommended biological properties. Characteristic of antigen, antibody, linker–antibody attachment, linker, linker–payload attachment and payload are detailed. The antibody Fc part is implied in half-life and immunogenicity. The Fab part controls affinity and avidity to the targeted antigen.
Figure 1. Design of an antibody–drug conjugate and recommended biological properties. Characteristic of antigen, antibody, linker–antibody attachment, linker, linker–payload attachment and payload are detailed. The antibody Fc part is implied in half-life and immunogenicity. The Fab part controls affinity and avidity to the targeted antigen.
Cancers 14 00778 g001
Figure 2. Overview of ADC mode of action. 1. Binding of the ADC to the antigen expressed by the cancer cell. 2. Internalization of the ADC. 3. Degradation of the linker or antibody inside the lysosome induces the release of an active form of the payload. 4. The payload exerts cellular toxicity depending on its mode of action. 5. A bystander effect can occur.
Figure 2. Overview of ADC mode of action. 1. Binding of the ADC to the antigen expressed by the cancer cell. 2. Internalization of the ADC. 3. Degradation of the linker or antibody inside the lysosome induces the release of an active form of the payload. 4. The payload exerts cellular toxicity depending on its mode of action. 5. A bystander effect can occur.
Cancers 14 00778 g002
Figure 3. ADC targets in oncodermatologic indications and implication of antigen expression in cancer aggressiveness. (A) Cutaneous T-cell lymphoma. (B) Melanoma. (C) Carcinoma: squamous cell carcinoma and Merkel cell carcinoma.
Figure 3. ADC targets in oncodermatologic indications and implication of antigen expression in cancer aggressiveness. (A) Cutaneous T-cell lymphoma. (B) Melanoma. (C) Carcinoma: squamous cell carcinoma and Merkel cell carcinoma.
Cancers 14 00778 g003
Table 1. FDA-approved ADCs. A. Hematological malignancies. B. Solid tumors.
Table 1. FDA-approved ADCs. A. Hematological malignancies. B. Solid tumors.
Hematological Malignancies
Commercial NameInternational Non-Proprietary Names (INN)TargetAntibody IsotypeBioconjugation Head (Antibody Amino Acid)LinkerDrug (Therapeutic Class)Indication
Adcetris®brentuximab vedotinCD30Chimeric IgG1Maleimidocaproyl (Cysteine)Cleavable/proteolytic (cathepsin B)auristatinanaplastic large cell lymphoma + Hodgkin’s lymphoma
Polivy®polatuzumab vedotin-piiqCD79bHumanized IgG1Maleimidocaproyl (Cysteine)Cleavable/proteolytic (cathepsin B)auristatinrelapsed or refractory diffuse large B-cell lymphoma
Mylotarg®gemtuzumab ozogamicinCD33Humanized IgG4Acetyl butyrate (Lysine)Cleavable/hydrazonecalicheamicinCD33-positive acute myeloid leukemia
Beponsa®inotuzumab ozogamicinCD22Humanized IgG4Acetyl butyrate (Lysine)Cleavable/hydrazonecalicheamicinlymphoblastic leukemia
Lumoxiti®moxetumomab pasudotoxCD22N/AN/ACleavable/proteolytic (furin)Pseudomonas endotoxin Ahairy cell leukemia
Zynlonta®loncastuximab tesirine-lpylCD19Humanized IgG1Maleimidocaproyl (Cysteine)Cleavable/proteolytic (cathepsin B)pyrrolobenzodiazepine dimer (PBD)relapsed/refractory diffuse large B-cell lymphoma
Blenrep®belantamab mafodotinCD38Humanized IgG1kMaleimidocaproyl (Cysteine)Uncleavableauristatinrelapsed/refractory multiple myeloma
Solid Tumors
Commercial NameInternational Non-Proprietary Names (INN)TargetAntibody IsotypeBioconjugation Head (Antibody Amino Acid)LinkerDrugIndication
Kadcyla®ado-trastuzumab emtansineHER2Humanized IgG1Maleimidocaproyl (Lysine)UncleavablemaytansineHER2/neu positive breast cancer
Padcev®enfortumab vedotinNectin-4Human IgG1Maleimidocaproyl (Cysteine)Cleavable/proteolytic (cathepsin B)auristatinlocally advanced or metastatic urothelial cancer
Enhertu®trastuzumab deruxtecanHER2Humanized IgG1Maleimidocaproyl (Cysteine)Cleavable/proteolytic (cathepsin B)deruxtecan (topoisomerase inhibitor)breast cancer HER2 positive after two or more lines of anti-HER2 therapy
Trodelvy®sacituzumab govitecanTROP-2Humanized IgG1Maleimidocaproyl (Cysteine)Cleavable/hydrazonetopoisomerase inhibitormetastatic triple-negative breast cancer
Tivdak®tisotumab vedotinTissue factorHuman IgG1Maleimidocaproyl (Cysteine)Cleavable/proteolytic (cathepsin B)auristatincervical cancer
Unprovided data: N/A.
Table 2. Preclinical and clinical trials of ADCs in cutaneous lymphoma.
Table 2. Preclinical and clinical trials of ADCs in cutaneous lymphoma.
Commercial NameTargetPercentage of PositivityExpression in Healthy TissuesAntibodyAntibody IsotypeBioconjugation Head (Antibody Amino Acid)LinkerDrugPhase
Brentuximab vedotinCD3075%Activated T, B and NK cellsBrentuximabChimeric IgG1Maleimidocaproyl (Cysteine)Cleavable/proteolytic (cathepsin B)MMAEApproved
F0002ADCCD3075%Activated T, B and NK cellsBrentuximabChimeric IgG1Ester NHS (Lysine)UncleavableDM1Phase I
N/AInducible co-stimulatorMF: 61% (n = 23), SS: 88% (n = 17)Lymph node, kidney, liverMogamulizumabMurine monoclonal antibodyN/AN/AMMAEPreclinical
N/ACell surface heat shock protein 70N/AN/A239-87N/AN/AN/AMMAEPreclinical
Unprovided data: N/A.
Table 3. Results of brentuximab vedotin clinical trials in cutaneous lymphoma.
Table 3. Results of brentuximab vedotin clinical trials in cutaneous lymphoma.
StudyPhasePatient NumberResponse RateSurvival
Duvic et al.II48 (28 with MF, 9 with LyP, 2 with pcALCL)ORR 73%
ORR 54% in MF group
ORR 100% in other subgroups
CR 2/28, PR 13/28 in MF subgroup
PFS 1.1 years (95% CI 0.9−1.4)
Kim et al.II32 (MF and SS)ORR in 21/30 (70%, 90% CI 53−83)
CR in 1/30
PR in 20/30SD in 4/30
Median PFS not reached at 12 months
Median EFS > 6 months
61% event free at 6 months
28% event free at 12 months
Prince et al.III
Comparison of BV with physician’s choice of either bexarotene or methotrexate
123 (97 with MF, 31 with pcALCL) with 64 in BV group, 64 in PC groupORR 56.3% (BV group) versus 12.5% (PC group), with p < 0.0001
ORR 67% in BV group
CR 16%
ORR 20% in PC group
CR 2%
Median PFS 16.7 months (BV group) versus 3.5 months (PC group), with p < 0.0001
Objective response rate: ORR; mycosis fungoides: MF; Sézary syndrome: SS; complete response: CR; partial response: PR; lymphomatoid papulosis: LyP; primary cutaneous anaplastic large cell lymphoma: pcALCL; progression-free survival: PFS; brentuximab vedotin: BV; physician’s choice: PC; event-free survival: EVS.
Table 4. Preclinical and clinical trials of ADCs in melanoma.
Table 4. Preclinical and clinical trials of ADCs in melanoma.
Commercial NameTargetPercentage of PositivityExpression in Healthy TissuesAntibodyAntibody IsotypeBioconjugation Head (Antibody Amino Acid)LinkerDrugPhase
Glembatumumab vedotingpNMB86% (n = 21)Skin, bonesGlembatumumabHuman IgG2Maleimidocaproyl (Cysteine)Cleavable/proteolyticMMAEPhase I/II
N/APMEL1764% (n = 58)Melanocytes17A9Mouse N/AN/ACleavable/proteolyticMMAEPreclinical
EV20/MMAF ADCHER365% (n = 130)Liver, pancreas, epithelial cellsEV20Humanized IgG1Maleimidocaproyl (Cysteine)UncleavableMMAFPreclinical
DEDN6526AETBRMajority of tumors (% N/A)Liver, cortex, medullaMEDN6000AHumanized IgG1Maleimidocaproyl (Cysteine)Cleavable/proteolyticMMAEPhase I
LOP628c-KIT66% to 88%Skin epithelial cells, breast, neuronsLMJHumanized IgG1Ester NHS (Lysine)UncleavableDM1Phase I (stopped)
Enapotamab vedotinAXLN/AMuscle, testisEnapotamabHuman IgG1Maleimidocaproyl (Cysteine)Cleavable/proteolyticMMAEPhase I/II
Unprovided data: N/A. gpNMB: glycoprotein-NMB; PMEL17: premelanosome protein 17; HER3: human epidermal growth factor receptor 3; ETBR: endothelin B receptor; c-KIT: tyrosine-protein kinase; AXL: Anexelekto.
Table 5. Preclinical and clinical trials of ADCs in carcinoma. A. Squamous cell carcinoma. B. Merkel cell carcinoma.
Table 5. Preclinical and clinical trials of ADCs in carcinoma. A. Squamous cell carcinoma. B. Merkel cell carcinoma.
Squamous Cell Carcinoma
Commercial NameTargetPercentage of PositivityExpression in Healthy TissuesAntibodyAntibody IsotypeBioconjugation Head (Antibody Amino Acid)LinkerDrugPhase
Tisotumab vedotinTF75% (n = 20)Brain, heart, intestine, kidney, lung, placenta, uterusTisotumabHuman IgG1Maleimidocaproyl (Cysteine)Cleavable/proteolyticMMAEPhase I/II
Bivatuzumab mertansineCD44v6100% (n = 5)Skin keratinocytes, cervix, cornea, tonsilBivatuzumab (or BIWA 4)Humanized IgG1Disulfide linker SPP (Lysine)Cleavable/hydrazoneDM1Phase I (stopped)
Samrotamab vedotin (ABBV-085)LRRC1564% (n = 115)Hair follicles, tonsil, stomach, spleen, osteoblastsAb1Humanized IgG1Maleimidocaproyl (Cysteine)Cleavable/proteolyticMMAEPhase I
Merkel Cell Carcinoma
Commercial NameTargetPercentage of PositivityExpression in Healthy TissuesAntibodyAntibody IsotypeBioconjugation Head (Antibody Amino Acid)LinkerDrugPhase
Lorvotuzumab mertansine (IMGN901)CD5688% (n = 64)NK cells, neuroendocrine cells, neuronsLorvotuzumab (huN901)Humanized IgG1Disulfide linker SPP (Lysine)Cleavable/hydrazoneDM1Phase I
Adcitmer®CD5666% (n = 90)NK cells, neuroendocrine cells, neuronsm906Human IgG1Maleimidocaproyl (Cysteine)Cleavable proteolytic (cathepsin B)MMAEPreclincal
TF: tissue factor; LRRC15: leucine-rich repeat containing 15.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Esnault, C.; Schrama, D.; Houben, R.; Guyétant, S.; Desgranges, A.; Martin, C.; Berthon, P.; Viaud-Massuard, M.-C.; Touzé, A.; Kervarrec, T.; et al. Antibody–Drug Conjugates as an Emerging Therapy in Oncodermatology. Cancers 2022, 14, 778. https://doi.org/10.3390/cancers14030778

AMA Style

Esnault C, Schrama D, Houben R, Guyétant S, Desgranges A, Martin C, Berthon P, Viaud-Massuard M-C, Touzé A, Kervarrec T, et al. Antibody–Drug Conjugates as an Emerging Therapy in Oncodermatology. Cancers. 2022; 14(3):778. https://doi.org/10.3390/cancers14030778

Chicago/Turabian Style

Esnault, Clara, David Schrama, Roland Houben, Serge Guyétant, Audrey Desgranges, Camille Martin, Patricia Berthon, Marie-Claude Viaud-Massuard, Antoine Touzé, Thibault Kervarrec, and et al. 2022. "Antibody–Drug Conjugates as an Emerging Therapy in Oncodermatology" Cancers 14, no. 3: 778. https://doi.org/10.3390/cancers14030778

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