Skip to main content

Current progress and remaining challenges of peptide–drug conjugates (PDCs): next generation of antibody-drug conjugates (ADCs)?

Abstract

Drug conjugates have emerged as a promising alternative delivery system designed to deliver an ultra-toxic payload directly to the target cancer cells, maximizing therapeutic efficacy while minimizing toxicity. Among these, antibody-drug conjugates (ADCs) have garnered significant attention from both academia and industry due to their great potential for cancer therapy. However, peptide-drug conjugates (PDCs) offer several advantages over ADCs, including more accessible industrial synthesis, versatile functionalization, high tissue penetration, and rapid clearance with low immunotoxicity. These factors position PDCs as up-and-coming drug candidates for future cancer therapy. Despite their potential, PDCs face challenges such as poor pharmacokinetic properties and low bioactivity, which hinder their clinical development. How to design PDCs to meet clinical needs is a big challenge and urgent to resolve. In this review, we first carefully analyzed the general consideration of successful PDC design learning from ADCs. Then, we summarised the basic functions of each component of a PDC construct, comprising of peptides, linkers and payloads. The peptides in PDCs were categorized into three types: tumor targeting peptides, cell penetrating peptide and self-assembling peptide. We then analyzed the potential of these peptides for drug delivery, such as overcoming drug resistance, controlling drug release and improving therapeutic efficacy with reduced non-specific toxicity. To better understand the potential druggability of PDCs, we discussed the pharmacokinetics of PDCs and also briefly introduced the current PDCs in clinical trials. Lastly, we discussed the future perspectives for the successful development of an oncology PDC. This review aimed to provide useful information for better construction of PDCs in future clinical applications.

Graphical abstract

Introduction

Chemotherapy remains the first-line treatment for various malignant cancers. However, its non-selective toxicity and acquired drug resistance pose significant challenges in clinical applications [1, 2]. To address these limitations, drug conjugates, comprising a tumor-targeting carrier, linker, and cytotoxic payloads, were designed to deliver toxic agents to tumor sites with increased therapeutic efficacy and lower side effects [3, 4]. As the most successful modality of drug conjugates, antibody-drug conjugates (ADCs) have been widely used for cancer treatments, exhibiting both high efficacy and favorable tolerability. Since the first ADC drug, mylotarg (gemtuzumab ozogamicin), was approved by the Food and Drug Administration (FDA) in 2000 for adults with acute myeloid leukemia (AML), ADCs have been attracting growing interest in the drug industry. By October 2024, 15 ADC drugs had been approved for hematological malignancies and solid tumors worldwide [5]. However, the high cost, low cellular penetration, undesired immunotoxicity, and emerging resistance undermine the development of ADCs [6]. The clinical limitation of ADCs has triggered various drug modalities for increased efficacy and lower toxicity. Nanobodies, single-domain antibodies, can deliver therapeutic drugs to target tissues with strong antigen-binding capacity and good tissue penetration, which have been evaluated in different stages of clinical trials for tumor diagnosis and therapeutics [7]. In recent years, the conjugation of nanobodies with functional moieties such as therapeutic drugs, radionuclides, and optical tracers has achieved specific tumor targeting, enhanced therapeutic efficacy, and reduced adverse effects [7,8,9]. Thus, the emergence of nanobodies could be a new drug modality that can overcome the limitation of ADCs.

Similar to ADCs, peptide-drug conjugates (PDCs) are defined as drugs covalently linked to peptides with certain functions via specialized linkers. PDCs can reach their target sites via homing peptides and release the toxic payload via linkers in targeted cells [10]. As a homing peptide, it must have specific targeting capabilities of protein receptors over-expressed at tumor tissues with strong binding affinity. The secondary structure of peptides plays a key role in the in vivo stability and affinity for targeted drug delivery [10]. As a linker, it must be stable in the circulation but efficiently release the toxic drug in target cells based on the specific tumor environment compared to normal cells such as low pH, over-expressed enzymes, et al. [11]. Meanwhile, the conjugation of PDCs should not affect peptide binding affinity and the toxicity of payloads. Understanding how PDCs work could help researchers design more efficient PDCs with good therapeutic efficiency and safety. PDCs are acknowledged to have many advantages over ADCs: (1) ease of synthesis by solid phase synthesis, liquid phase synthesis, or biosynthesis; (2) facile functionalization via different chemical modifications, such as cyclization, N-methylation, unnatural amino acid incorporation; (3) high tissue penetration; (4) low immunotoxicity; (5) overcoming drug resistance in tumor cells by altering the cell entry mechanism of small drugs [12, 13]. As of 2020, There were > 100 approved peptides with therapeutic or diagnostic applications, and hundreds of peptides are under clinical trials for the treatment of cancer, infection, diabetes, pain treatment, et al. [10]. Witnessing the success of peptides, PDCs, are now attracting increasing interest in drug discovery. However, PDCs have progressed slowly in clinical application since they were first reported in 1972 by Freer et al. [14]. Only several radioactive PDCs are approved for cancer diagnosis and therapy, such as 68Ga-DOTATOC, 68Ga-DOTATATE, 177Lu-DOTATATE [15]. What possible barriers and challenges do PDCs meet when applying in the clinic? This review will first highlight the general consideration of successful PDC design learning from ADCs. Then, the basic function and choices of each component of a PDC construct will be summarized. The status of PDC clinical development will be introduced in the following section. Lastly, the future perspectives for the successful development of an oncology PDC will be discussed and concluded. The purpose of the review is to provide guidance for well construction of peptide-drug conjugates for clinical application.

General consideration of PDCs learning from ADCs

With the rapid progress of ADC candidates in clinical applications, major challenges such as undesired toxicity and drug resistance have also drawn much attention. The experiences of ADC construction are worth learning to circumvent the pitfalls encountered with ADCs. Before introducing the general consideration of PDCs from ADCs, we compared the properties of ADCs and PDCs, which could help better understand their differences, as shown in Table 1 [16]. As ADCs met a range of limitations, in the following section we will discuss some pitfalls of traditional ADC therapeutics and highlight ways that PDCs can overcome these challenges, concluded in Fig. 1 and the following introductions of PDCs.

Table 1 The comparison of PDC and ADCs

Off-site toxicity

A previous study showed that out of 79 ADC development programs terminated since 2000, 32 were discontinued due to safety concerns [20]. Therefore, effectively managing toxicity and enhancing safety are critical factors for developing ADCs successfully. The off-site toxicity of ADCs can be categorized into “on-target toxicity” and “off-target toxicity.” On-target toxicity occurs when ADC-mediated targeted uptake affects normal tissues expressing the same target. Off-target toxicity is caused by the pre-release in the circulation and non-specific uptake of payloads. The key to solving this problem of “On-target toxicity” is exploring novel targets expressed exclusively or predominantly in tumor cells but rarely or low in normal tissues. The optimization of targeting units to the selected targets is also crucial for a successful drug conjugate. Peptide ligands come from natural plants, animals, chemical synthesis, or de novo discovery approach. The rapid progress of peptide screening technologies accelerated the discovery of specific peptide ligands with high receptor affinity, including phase display [21], one-bead-one-compound (OBOC) method [22], mRNA display [23], split-intein circular ligation of peptides and proteins (SICLOPPS) et al. [24]. Compared to mAbs, peptides had lower molecular weight to overcome the drawback of low penetration along with rapid clearance from circulation. Peptide stabilization strategies have been widely developed to improve their physiological stability and pharmacokinetics, such as peptide stapling [25] and non-basic amino acid substitution [26, 27].

Thus, with this advanced technology in hand, the rapid identification of drug targets will greatly boost the development and clinical translation of drug conjugates with high efficiency and low toxicity.

Fig. 1
figure 1

The challenges of ADCs and the chances for PDC in future drug development

The limited choice of payloads

In addition to on-target toxicity, bystander effects arise from non-target-mediated cellular uptake and catabolism in normal cells, leading to the release of the cytotoxic payload and/or its metabolites. This off-target toxicity is particularly evident in payload-associated platform toxicities, where ADCs utilizing the same linker-payload combination exhibit similar safety profiles, regardless of the target antigen [28]. Recent studies revealed that only 1–2% of ADC payloads reach the intracellular target. Thus, the potency of the payload must be high enough to eradicate tumor cells even at a low concentration [29]. However, the highly potent payloads might lead to severe side effects, especially a significant level of antigen expression on critical normal cells. Compared to ADCs, PDCs had more payload choices. Besides radionuclides and chemo-agents, toxic proteins, cytokines, PROTACs, and oligonucleotides could also be designed for the next generation of therapeutic PDC [30].

Drug resistance

The mechanisms of drug resistance are complicated and associated with enhanced efflux of drugs, elevated metabolism of xenobiotics, signaling pathway compensation, target change, cell death inhibition, et al. [31,32,33]. As for ADCs, efforts to overcome the resistance included: (1) choice of the payloads that are poor efflux substrates, such as trastuzumab deruxtecan, using a novel DNA topoisomerase I inhibitor, can overcome T-DM1 resistance caused by aberrant expression of ATP-binding cassette (ABC) transporters in HER2-positive gastric cancer [34]; (2) increasing the hydrophilicity via linker modification as MDR1 transports hydrophobic compounds more efficiently than hydrophilic compounds [35]; (3) developing of bispecific/ biparatopic ADCs [36]. These strategies could also be used for PDC construction. Moreover, cell penetration peptides could change the cellular uptake mechanism of chemo-agents, which could be an alternative method to overcome drug resistance. Meanwhile, due to the facile modification of peptides, two different drugs could be conjugated to peptides for combination therapy and avoiding drug resistance.

To sum up, the rapid development of ADCs also meets some limitations or challenges for their wide clinical application. The progress in PDCs is still in the early stages, but they have promising potential as they have unique advantages for targeted drug delivery. A better understanding of the design of ADCs and the components of PDCs could be beneficial for future PDCs’ construction.

Key components of PDCs

PDCs are an emerging class of prodrugs formed through the covalent attachment of a specific peptide sequence to a drug via a cleavable linker. The diversity and facile construction of peptides allow for incorporating a great degree of functionality into PDCs, such as enhancing tumor targeting of chem-agents, circumventing multiple drug resistance, inducing self-assembling for drug delivery, and so on [37]. Besides peptide diversity, linker structure also plays a key role in manipulating the biological action of PDCs in different manners, for example, the cleavable linkers can be triggered by tumor-specific microenvironments such as pH, tripeptide glutathione (GSH) or enzymes to reach a tumor-targeted therapy, while the non-cleavable linkers can be stable in the circulation avoiding the release of payloads in blood and reduction of anti-cancer effect [38]. The payload selection is also important for PDC construction and application. In this part, we will systematically introduce the role of each component and how alternative approaches can improve the current limitations of this emerging modality.

Peptides in PDCs

Similar to antibodies, peptides are composed of amino acids linked together by peptide bonds. However, peptides have lower molecular weights and generally lack the complex tertiary structures found in antibodies. Given their ubiquitous presence in biological systems and their involvement in regulating various physiological processes, peptides provide an alternative approach to develop PDCs. The peptides significantly impact efficacy, pharmacokinetic/pharmacodynamic profile, and therapeutic index of PDCs as their influence on tumor targeting, in vivo stability, and cell penetration properties. Unlike mAbs, peptides can act as homing peptides for tumor targeting and can also be used as cell penetration enhancers to transport small drugs into cells. According to the cellular function, peptides in PDCs can be divided into tumor-targeting peptides, brain penetration peptides, cell penetration peptides, and self-assembling peptides (Fig. 2), which will be discussed in this part [10, 13, 39, 40].

Fig. 2
figure 2

Multifunctional peptides used in PDCs, including cell penetration peptides, tumor targeting peptides, and self-assembling peptides

Tumor-targeting peptides

Tumor-targeting peptides have become an increasingly useful tool for targeted delivery of therapeutic and diagnostic agents into tumors. The ideal peptide for PDCs should have a strong target binding affinity, high selectivity to tumor cells, and in vivo stability. Herein, we focused on several potential tumor-targeting peptides used in PDCs and discussed their behaviors in delivering chemo-agents to tumor tissues, which helped distinguish the proper homing peptide for clinical application. To make this part comprehensive, we categorize the targets according to their function, including targeting immune checkpoint, targeting tumor vasculature, targeting hormone-associated receptors, targeting the tumor microenvironment, targeting the blood-brain barrier (BBB), and other targets (Table 2).

Table 2 Classification of tumor-targeting peptide in PDCs

Anti-angiogenic targets

Integrins Ανβ3

The tripeptide motif arginine-glycine-aspartic acid (RGD), identified in the 1980s within fibronectin, could specifically bind to integrins ανβ3 and ανβ5 and was frequently used as a peptide carrier for targeted cancer therapy [41]. The cyclic RGD peptide cyclo (RGDfK), which has good stability and target affinity, has gained considerable attention for different kinds of drug conjugation. Several radionuclide-RGD conjugates have entered clinical studies as positron emission tomography (PET) tracers including [18F] fluciclatide [41], [18F]RGD-K5 [42], 68Ga-NOTA-bombesin(BBN)-RGD et al. [43]. However, despite the advantages of the RGD motif, cilengitide was discontinued in 2014, and three Phase II clinical trials of [18F] fluciclatide have been withdrawn. In 2023, 68Ga-FAPI-RGD with the conjugation of fibroblast activation protein (FAP) inhibitors (FAPIs) and RGD have been evaluated in patients with FAP- and integrin αvβ3-positive tumors. The clinical results demonstrated an improved lesion detection rate and tumor delineation, particularly for the diagnosis of lymph node (99% vs. 91%) and bone (100% vs. 80%) metastases compared to 18F-FDG PET/CT (NCT37142301) [44]. Studies also revealed that RGD peptide can efficiently deliver chemo-agents to the tumor tissues with enhanced tumor accumulation and anti-cancer effect, including doxorubicin (DOX) [45], camptothecin [46], paclitaxel (PTX) [47], platinum (Pt) [48]. Despite the advantages of the RGD motif, cell biology research revealed that the combination of RGD with RGD-positive cells will induce cell deformation, which is conducive to the attachment, spreading, and migration of RGD-positive cells. Thus, when RGD is used as an integrin-specific binding ligand, it is necessary to fully consider the possibility of inducing the spread of tumor cells.

Mammalian aminopeptidase N (APN) (or CD13)

Asn-Gly-Arg (NGR) motifs have vasculature-homing properties via interactions with the aminopeptidase N (CD13) expressed on tumor neovasculature [49]. Many NGR-based conjugates have been reported for cancer therapy or imagine, such as 99mTc-labeled NGR-chlorambucil conjugate [49], 99m Tc-HYNIC-CLB-c(NGR) [49], cyclic NGR peptide-daunomycin conjugates [50], RGD-NGR-NSAID(Non-steroidal anti-inflammatory drug) conjugates [51]. Most NGR-drug conjugates showed limited selectivity to CD13-positive cancer cells. Currently, only two fusion proteins conjugated with homing peptides are in clinical trials: NGR-human tumor necrosis factor (hTNF) and truncated tissue factor (tTF)-NGR [52]. NGR-hTNF was awarded “orphan drug status” for malignant pleural mesothelioma and hepatocellular carcinoma in the EU and the USA and is now in a Phase I clinical trial for cancer patients with solid tumors or lymphomas. The (tTF)-NGR targeting tissue factor to tumor vasculature can induce tumor infarction, which was reported to inhibit solid tumor growth in mice. Now, tTF-NGR is in a Phase I clinical trial for cancer patients with solid tumors or lymphomas [53].

CD133

CD133 is a transmembrane glycoprotein over-expressed in various solid tumors, which is not only the most prominent cancer stem cell (CSC) marker but also contributes to neovascularization in vivo [85, 86]. Sun et al. discovered a CD133 targeting linear peptide LS-7 (LQNAPRS) using phage display technology with good target affinity and selectivity [87]. Based on this study, Tao et al. designed a novel PDC by conjugating camptothecin (CPT) with peptide LS-7 via a succinyl linker [54]. This PDC could overcome the limitation of CPT with poor solubility and high systemic toxicity, which demonstrated improved biodistribution, good therapeutic activity, and a better safety profile in vivo. This discovery provides a new approach for the delivery of toxic CPT to targeted tumor tissues for the therapy of cancer stem-like cells.

VEGFR

Vascular endothelial growth factor (VEGF) is a well-characterized angiogenic factor that plays an important role in tumor-associated angiogenesis, tumor recurrence, and metastasis by binding to their receptors VEGFRs. Wang et al. discovered a VEGFR3 targeting peptide CP7 (CIQPFYP) by computational design, which exhibited high VEGFR3 targeting affinity and tumor-homing efficiency [88]. Then, they conjugated this homing peptide to PEG-b-PLL polymers to enhance the tumor accumulation and chemotherapeutic efficacy of DOX [89]. As linear peptide ligands could readily degrade, Ying et al. discovered a cyclic A7R peptide(cA7R) by computer-aided peptide design and head-to-tail cyclization [90]. This cyclic peptide ligand could enhance the delivery of doxorubicin (DOX) loaded liposomes to tumor tissues. Besides conjugated nanoparticles, VEGFR targeting PDCs were also discovered by us through the conjugation of VEGFR targeting peptide VEGF125 − 136 (QKRKRKKSRYKS) to a lytic peptide. This PDC could both suppress tumor angiogenesis and tumor proliferation in a TACE model for hepatocellular carcinoma therapy [55].

Prostate-specific membrane antigen (PSMA)

Prostate-specific membrane antigen (PSMA) is a protease over-expressed in prostate cancer cells and the endothelium of tumor vasculature but much less so in normal tissues [56]. PSMA processes PSMA-specific peptide substrate (Asp-γ-Glu-γ-Glu-γ-GluGluOH) is processed by PSMA to provide an analog that is now cell-permeable, cytotoxic, and extracellularly concentrated adjacent to cancerous prostate cells. G202, consisting of a PMSA-targeting peptide substrate and cytotoxic thapsigargin, was hypothesized to be effectively and efficiently cleaved by the extracellular enzymatic activity of PSMA, thereby delivering cytotoxic agents specifically to a tumor site. Nowadays, G202 has been entered into several Phase 2 clinical trials, including prostate cancer, renal cell carcinoma, hepatocellular carcinoma, and glioblastoma, but clinical results have not yet been reported [57].

Immune checkpoint targets

PD-L1

Immune checkpoint blockade (ICB) has provided significant clinical advances in various cancer types via monoclonal antibodies against programmed cell death protein 1 (PD-1), programmed death-ligand 1 (PD-L1), or cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) [91]. PD-L1 is overexpressed in cancer cells and plays a major role in suppressing T-cell activation. As the potential ability of PD-L1 on tumor progression, many PD-1/PD-L1 peptide antagonists had been rapidly developed and used as warheads for targeted drug delivery [92,93,94]. PDCs targeting PD-L1 are also designed and applied for cancer immunotherapy. For instance, Kim et al. developed peptide-oligonucleotide conjugate (Pep-21) consisting of a PD-L1-binding peptide (NYSKPTDRQYHF) covalently linked with an anti-miR-21 inhibitor via click chemistry, which was preferentially internalized in both cancer cells and tumor-associated macrophages (TAMs) [58]. This conjugate exhibited reduced tumor cell migration and restrained tumor progression. A shorter PD-L1 peptide(CVRARTR) used for PDC construction also showed a potent anti-cancer effect. This conjugate consists of an anti-PD-L1 peptide, cathepsin B-specific cleavable peptide (FRRG), and doxorubicin (DOX) and can be self-assembled into nanoparticles via intermolecular interactions [59]. This drug delivery system can achieve targeted tumor delivery of anti-PD-L1 peptide and DOX and efficiently inhibit tumor progression with minimal side effects. Zeng et al. designed a new PD-L1 degrader by conjugation of PMT degraders with an anti-PD-L1 peptide featuring disulfide linkers This conjugate achieved synergistic immunotherapeutic effect via both external PD-L1 blockade and internal PD-L1 degradation [93].

CD47

CD47 is over-expressed in many cancer types and induces immune evasion by sending out signals like “don’t eat me” via the CD47-SIRP α-axis [95]. CD47 has been an attractive target for cancer immunotherapy. In recent years, CD47 targeting peptides have been developed to inhibit CD47-SIRP α-axis, such as RS17 (RRYKQDGGWSHWSPWSS) [60]. As for PDC design, Pan et al. constructed a CD47 by conjugation of CD47-blocking peptide VK17 with pro-apoptotic VK30 peptide. This PDC can induce lung cancer cell apoptosis and macrophage phagocytosis and suppress lung cancer cell growth in mice. Another CD47 targeting peptide (CkRFYVVMWKk, k = D analog of lysine) was discovered to deliver gemcitabine to bladder cancer (BC) cells over-expressed with CD47 with high tumor accumulation [61]. Thus, the inhibition of the CD47-SIRPαinteraction by peptides or PDCs is a potential approach to cancer immunotherapy.

Hormone associated targets

SST2 receptor

Peptide hormone-based targeted therapy has been studied extensively. As a somatostatin receptor, the SST2 receptor subtype is highly expressed in various tumor cells and primary tumor tissues and has become an attractive target for SST receptor-mediated anti-cancer therapy [62]. Octreotide is a somatostatin analog with a high affinity for the SST2 receptor [62]. Octreotide-derived delivery strategies have been proposed and explored for tumor-targeted therapeutics or diagnostics in preclinical or clinical settings. Octreotide-based radioactive conjugates have been applied clinically to detect small neuroendocrine tumor sites, including 111In-DTPA-octreotide with high renal clearance and less intestinal accumulation [63], 99mTc-HYNIC/EDDA-3Tyr-octreotide for the diagnosis of neuroendocrine tumors [64], 68Ga-DOTATOC, and 68Ga-DOTATATE [65] et al. for gastroenteropancreatic neuroendocrine tumors. Meanwhile, Octreotide-small drug conjugates have also been designed, such as doxorubicin [66], paclitaxel [67], and camptothecin [68].

GnRH receptor

GnRH receptor, or LH-RH (Luteinizing hormone-releasing hormone) receptor, is expressed in many endocrine cancers, including breast, ovarian, endometrial, and prostate tumors [69]. Zoptarelin Doxorubicin (AN-152, AEZS-108, ZoptrexTM) is a peptide-drug conjugate composed of a GnRH analog ([D-Lys(6)]-GnRH) and doxorubicin The conjugate proved more effective than doxorubicin in inhibiting cell proliferation in GnRH receptor-positive cancer cell lines and several xenograft mouse tumor models. Meanwhile, this PDC successfully entered several clinical trials, which we discussed in the below part [69,70,71,72]. Besides chemo agents, an antiangiogenic compound (sunitinib) can be conjugated to the targeting peptide [D-Lys(6)]-GnRH, generating SAN1GSC, which had significant tumor growth delay compared with equimolar sunitinib alone in castration-resistant prostate cancer animal models [73].

Tumor microenvironment targets

Matrix metalloproteinase (MT1-MMP)

Overexpression of membrane type-1 matrix metalloproteinase (MT1-MMP) is closely associated with malignancies such as breast, lung, ovarian, and colon cancers [74]. BT1718 is a novel bicyclic peptide-drug conjugate targeting MT1-MMP to release its toxic payload DM1 [74, 75]. The bicyclic peptide binding to MT1-MMP is identified by phage display technology as the tumor-target peptide. The bicyclic peptide also has other inherent characteristics to minimize drug toxicity, such as short systemic half-life, rapid tumor penetration, low systemic exposure, and rapid renal clearance.

Blood-brain barrier targets

Low-density lipoprotein receptor-related protein 1 (LRP1)

LRP1 has been reported to be expressed on the blood-brain barrier (BBB) and to be responsible for the transportation of multiple ligands across the BBB [76]. ANG1005, the most typical PDCs targeting LRP1, has been reported to deliver paclitaxel to cross the blood-brain barrier for the treatment of metastatic breast cancer in clinical trials, which showed notable CNS and systemic treatment effects in all treated patients [77]. To treat primary and secondary brain cancers, two PDCs named ANG1007 and ANG1009, consisting of three doxorubicin molecules conjugated to Angiopep-2, exhibited dramatically higher BBB influx rate constants than unconjugated doxorubicin, demonstrating their potential on the treatment of brain cancers [78].

Other targets

Hepatocyte receptor A2 (EphA2)

Erythropoietin-producing hepatocellular receptor A2 (EphA2) is a receptor tyrosine kinase family member that regulates cell-cell interactions such as differentiation, adhesion, migration, and death [79, 80]. EphA2 is overexpressed in a variety of solid tumors and is related to poor prognosis in cancer patients. BT5528 is a bicyclic peptide drug conjugate, which comprises a bicyclic peptide as a highly active EphA2 ligand and toxic auristatin E. BT5528 can accumulate in tumor tissues with a very low plasma concentration, which increases the selectivity for killing tumor cells with low systematic toxicity [80]. YSA (YSAYPDSVPMMS) is another EphA2-targeting peptide identified by phage display that effectively delivers anti-cancer agents to cancer tumors. For instance, YSA-paclitaxel conjugate can target EphA2 over-expressing prostate cancer cells in vivo, inhibiting tumor growth in a prostate cancer xenograft model more effectively than paclitaxel alone [81].

In addition to the introduced targets, several other peptide receptors are also investigated as potential targets for anti-cancer drug delivery. This target included epidermal growth factor receptor (EGFR) [82,83,84], human Y1 receptor (hY1R) [96] et al., and these receptors were overexpressed in specific tumor cell surfaces, and the detailed information was concluded in Table 2.

Cell penetrating peptides(CPP)

The cell membrane poses a barrier that limits the transport of macromolecules, proteins, nucleic acids, and drugs, restricting their penetration. Therefore, developing drugs capable of traversing cancer cell membranes is critical to achieve intracellular destruction. CPPs, ranging from 5 to 30 amino acid residues, are rich in arginine and lysine, which carry positive charges and exhibit amphiphilic properties in a physiological environment [97]. These features make CPPs effective for delivering otherwise cell-impermeable compounds or drugs into cells [98]. The widely recognized mechanisms by which CPP penetrates cell membranes can be divided into two categories: endocytosis and direct translocation, and the penetration mechanisms could be influenced by the chemical structure of peptides [99, 100]. CPPs in PDCs can be used to (1) improve the solubility and toxicity of small drugs, (2) change the drug cell penetration mechanism to overcome drug resistance, and (3) reduce the non-specific toxicity of small drugs.

Improving anti-cancer effects

Chen et al. constructed three PDCs (NTD, d-NTD, and q-NTD) in which TAT peptide is covalently attached to one, two, or four doxorubicin. It was shown that q-NTD had the most potent anti-cancer effect with the slowest drug release efficiency, indicating that the efficiency of intracellular accumulation and the rate of free drug release are essential factors in predicting the efficacy of PDCs in vitro [101]. Additionally, CPPs have been designed to target lymphatic metastases via intravenous injection. For instance, nanoparticles modified with CPP have demonstrated a 63.3% inhibition rate of lymphatic metastasis in lung cancer and reduced tumor growth rates by 1.4-fold [102].

Overcoming drug resistance

Well-designed PDCs can also greatly assist in overcoming the emergence of multidrug resistance (MDR) in tumors. For example, one particular mechanism of drug resistance is the increased expression of membrane proteins that mediate drug efflux, such as P-glycoprotein (Pgp), while guanidinium-rich PDCs can help circumvent these efflux proteins as their internalization does not involve passive diffusion across the membrane [103, 104]. For example, Wender et al. conjugated paclitaxel (PTX) to an octaarginine peptide transporter with significantly improved activity against malignant cells that were resistant to the PTX alone [105].

Reducing non-specific toxicity

An activatable PDC with cell-penetrating peptide and DOX conjugation (DOX-CR8G3PK6-DMA) was designed to achieve tumor-targeted drug delivery and reduce the non-specific toxicity of small drugs [106]. At the tumor extracellular pH of 6.8, hydrolysis of DMA occurred, and the cell-penetrating function of R8 was recovered to enhance cellular uptake of DOX with decreased systematic toxicity. Besides reducing the non-specific toxicity of chemo-agents, we recently designed cyclic PDC conjugated with pan-HDAC inhibitor SAHA, shown in Fig. 3, which is the cyclic peptide derived from HDAC1-specific substrate H3K56 [107]. This PDC showed selective toxicity to HDAC1-sensitive cell lines but poor toxicity to normal cancer cells. Then, we also developed sulfonium-tethered lytic peptides to conjugate with SAHA, exhibiting a selective anti-cancer effect [108]. Despite the versatility of CPPs used for PDCs and the fact that these conjugates would be a valuable tool for the treatment of cancers, their highly charged nature poses problems for general application and translation into the clinic.

Fig. 3
figure 3

Stabilized peptide drug conjugate targeting HDACs for cancer therapy. (A) The design of PDCs targeting HDAC. H3K56 peptide is derived from Histone 3, which is the substrate of HDAC1. The functional group hydroximic acid plays a key role for HDAC binding [107]. (B) Sulfonium-tethered PDCs targeting HDAC with decreased non-specific toxicity [108]. Copyright permission obtained with ref. no.83–84 from the author

Self-assembling peptide

The hydrophilic peptides combined with small hydrophobic drugs can observe overall amphiphilicity, introducing a tendency for self-aggregation in aqueous environments. The self-assembling PDCs can induce the formation of hydrogels or supramolecular nanostructures and are now receiving rapidly growing interest in the drug delivery community.

Controlling drug release

One important application of these self-assemblies is the sustaining drug release. The Stupp group has developed a range of peptide amphiphile (PA)-based hydrogels for biomedical applications [109, 110]. In one example, they conjugated the anti-inflammatory drug nabumetone to peptide amphiphiles through hydrazone bonds, and this conjugate could form a gel that release nabumetone in the low pH environment of the tumor [111]. The self-assembling PDCs can be used for the tumor-selective drug release activated by the tumor environment. For instance, the Xu group used the enzyme-instructed self-assembly strategy (EISA) to form nanofiber networks via a phosphorylated naphthalene-Phe-Phe-Lys-Tyr precursor conjugated with taxol [112]. Upon treatment with alkaline phosphatase (ALP) over-expressed in the tumor environment, the phosphate group was cleaved, and a translucent hydrogel spontaneously formed and exhibited comparable anti-cancer abilities with free taxol. To further improve the bio-stability, the Xu group developed D-amino acids-based hydrogels and could sustainably release free taxol for up to 1 month, leading to significant suppression of tumor growth [113].

Increasing drug loading and efficacy

Well-designed PDCs could also form nanostructures to provide targeted drug delivery and release. Cui et al. designed a novel class of PDCs that can form nanostructures by covalently conjugating one or more anti-cancer drugs to a rationally designed peptide through a biodegradable linker with a high drug loading and stability [114, 115]. Meanwhile, the positively charged peptide drug conjugates can also be self-assembled with negatively charged nucleic acid such as DNA or siRNA. We previously developed the HDAC-targeted PDC by the conjugation with a positive CPP and HDACi, which was then induced to be self-assembly by the interaction with anti-cancer DNA aptamer AS1411 [116]. This nanoparticle showed a synergetic anti-cancer effect through multiple signal pathways with low non-specific toxicity (Fig. 4).

Fig. 4
figure 4

An assembly-inducing PDC deliver nucleic acid for cancer therapy. This PDC consists of the cell penetration peptide RW9, an HDAC inhibitor warhead (peptide C-terminus), and 5-FU (peptide N-terminus), which can coassemble with AS1411 to form nanospheres. This PDC showed unexpected synergy with AS1411 to augment the cancer cell suppression efficiency, exemplified by the downregulation of the stemness-related proteins and the upregulation of apoptosis-related proteins. Copyright permission obtained with ref. no 92 from The Royal Society of Chemistry

Improving drug bioavailability

Peptide polymers are biodegradable polymers that can be conveniently synthesized and widely used for conjugated drug delivery [117]. The polypeptides–drug conjugates for anti-cancer therapy had been systematically introduced by Chen et al. [117]. For instance, polypeptides-chemotherapeutic drug conjugates have been developed to improve the bioavailability, enhance the therapeutic effects, and reduce the systemic toxicity of chemotherapeutic drugs. Kataoka et al. first conjugated DOX to the side chains of PEG-b-PAsp by an acid-labile hydrazone (Hyd) bond that can be cleaved in the endolysosomes with an acidic pH of 5–6, which presented intracellular acidity-triggered DOX release behaviors, tumor-infiltrating permeability, and effective anti-cancer activity with extremely low toxicity [118].

Despite the self-assembling PDCs showing good potential for targeted drug delivery, they also meet many challenges before they enter the clinic, such as possible structural instability during self-assembly, unpredictability of drug release, and possible non-specific toxicity to normal cells. The key step to solving these problems is understanding exactly how observed structures are formed. To study the formation process, Cai et al. carried out a Brownian dynamics (BD) simulation on a model rod-coil block copolymer/rigid homopolymer binary system, which helped control the ordered structures of the self-assembly [119, 120]. To address the challenge of drug release behaviour of the self-assembly, Buxton and co-workers developed a new simulation method to simulate the drug release performance of nanoparticles and observed an increased drug release rate near tumors [121]. These simulation works provide molecular-level insight into drug delivery behaviors, which greatly benefits the design of desirable drug delivery systems. In recent years, Matthew et al. explored the specific contributions of the drug and its linker to PDC self-assembly, which provided design principles for the enhanced control of PDC nanomaterials as therapeutic drug carriers [122]. To sum up, with a better understanding of the mechanisms of the formation process and the contribution of peptides, linkers, and drug molecules in self-assembly, there will be good drug candidates suitable for clinical applications.

Linkers

In PDCs, the linker connects the peptide to the drug, playing a crucial role in preventing off-target or peripheral drug release and enhancing the efficacy of the PDC. A well-designed linker should reduce the drug’s activity during delivery to minimize off-target toxicity and release the drug fully active upon reaching the target cells. Also, the bond between the peptide and the linker should not influence the affinity of the peptide for its receptor. Based on their behaviors after cellular uptake or under in vivo conditions, these linkers can be broadly divided into four groups: enzyme-cleavable, acid-cleavable, reducible disulfide, and non-cleavable (Fig. 5 and Table 3). Kaur et al. gave a detailed review of all aspects of linkers used in the design of PDCs [11] (Fig. 6).

Fig. 5
figure 5

Different linkers used in PDCs. The linkers used in PDCs included cleavable linkers and non-cleavable linkers

Table 3 Classification of different linkers used in PDC
Fig. 6
figure 6

The design of VEGFR targeting peptide conjugate (QR-KLU) and the antineoplastic efficacy of peptide QR-KLU in vitro and in vivo. Peptide QR-KLU is a conjugation of VEGFR targeting peptide VEGF125 − 136 (QKRKRKKSRYKS) and a reported lytic peptide(KLUKLUKKLUKLUK). This PDC showed good in vivo anti-tumor effect than traditional drug DOX through TACE in VX2 rabbit tumor model, and efficiently inhibited angiogenesis in tumor tissues with good safety. Copyright permission obtained with ref. no [55]. from Springer nature

Enzyme cleavable linker

Enzyme cleavable linkers are popular choices for both ADCs and PDCs, as they can be selectively cleaved by enzymes over-expressed in the tumor microenvironment or the lysosomes, such as matrix metalloproteinases(MMP), cathepsin B [38, 123]. Studies revealed that some of the enzymes are usually inactive outside the cell due to environmental pH, which is beneficial for their stability in the circulation. MMP2 and MMP9, members of MMPs, are essential in metastatic tumor cells, facilitating tumor development and angiogenesis. You et al. developed a unique human epidermal growth factor receptor 2 (HER2) targeting PDC via an MMP-2 sensitive linker (PVGLIG) to conjugate the trastuzumab-derived peptide with DOX [123]. This PDC not only had a more potent in vivo anti-cancer effect but also had a longer half-life (17.6 h versus 7.7 h) and slower clearance compared to free DOX. To further facilitate the release of active drugs from the PDC, specific dipeptide or tripeptide sequences have been employed. The dipeptide (Val-Cit or VC) can be specifically cleavage by cathepsin B and used for cRGD and DOX conjugation, which displayed superior cellular uptake, cytotoxicity toward integrin αvβ3 overexpressing B16 cells in vivo compared to another PDC containing disulfide linker [38]. Another tripeptide sequence is the Ala-Ala-Asn (AAN) cleaved by enzyme legumain overexpressed in tumor cells and microenvironment [124]. This tripeptide was widely used for the design of prodrugs rather than PDCs, which could significantly reduce the non-specific toxicity of these prodrugs. However, the instability of such short peptide linkers could lead to the release of drugs in pre-clinical in vivo studies.

Other enzymes rich in intracellular compartments, like endosomes and lysosomes of cancer cells, are esterases and amidases [125,126,127]. Karampelas et al. designed a PDC utilizing an ester bond and an amide bond to attach gemcitabine and peptide ligand targeting gonadotropin-releasing hormone-receptor (GnRH-R) [128]. As expected, this PDC showed a significant anti-cancer effect on a GnRH-R-positive prostate cancer xenograft animal model compared to gemcitabine. However, pharmacokinetic studies revealed that this PDC administration in mice resulted in the presence of high levels of gemcitabine, suggesting the rapid cleavage of the conjugate at the ester site, releasing the free drug from the peptide-linker moiety.

A carbamate linker is a stable linker used for PDC construction. Sayyad et al. explored the use of a carbamate linker in the design of PDC and discovered that the carbamate linker was more stable in vivo and released the drug slowly compared to the PDCs with an ester linkage, resulting in less toxicity to cancer cells [129]. However, it is unclear which linker chemistry will lead to better efficacy for tumor reduction. Based on many studies, the order of in vivo linker stability from greatest to least is as follows: amide > carbamate > ester > carbonate.

Recently, Kim et al. constructed an albumin-binding caspase-3 cleavable peptide-drug conjugate by conjugating doxorubicin to KGDEVD peptide on an albumin-binding moiety [130]. The increased albumin metabolism could induce cancer cell apoptosis, and the upregulation of caspase-3 could cleave the DEVE peptide and release DOX to exert a potent anti-cancer effect. This discovery provides a promising method for the treatment of pan-KRAC mutant cancer cells. Later, they designed another albumin-binding peptide-docetaxel conjugate platform (MPD3) with this caspase-3 cleavable liner [131]. This PDC showed good tumoral accumulation of cytotoxic payloads and robust anti-tumor activities in both local and liver metastatic PDAC tumor models in mice. These results proved the efficacy of caspase-3 cleavable linker in PDC construction.

Acid cleavable linkers

The acid-labile bonds usually maintain stability in blood circulation (pH7.4) but get cleaved primarily in the acidic tumor microenvironment (pH 6.5 − 6.9) or the acidic cellular compartments, endosomes (pH 5.5 − 6.2) and lysosomes (pH 4.5-5.0), including acetals, imines, hydrazine and various metal-organic frameworks (MOFs) [132,133,134,135]. Hydrazone is a popular acid-sensitive linker utilized for controlled drug release, and it has been successfully used in an FDA-approved ADC gemtuzumb ozogamicin (Mylotarg) [132], for PDCs, most studies used this linker to conjugate a doxorubicin (DOX) with homing peptides. For example, a hybrid 24-mer peptide was linked with a DOX via the hydrazone linkage, in which the hybrid peptide consisted of an ERK peptide inhibitor and a peptide ligand of transferrin receptor (TfR) over-expressed in the tumor cell [136]. This PDC showed a more obvious in vivo anti-cancer effect and higher distribution in the mouse tumor compared to the free Dox-treated mice. However, another PDC containing triple-negative breast cancer (TNBC) cell targeting peptide and a DOX via hydrazone linker was not stable in media and human serum with a half-life of 6 h but had TNBC selective toxicity compared with normal cells [133]. To improve the in vivo application of these conjugates, Yousef pour et al. synthesized a PDC (20 or ABD-Dox) using aldoxorubicin and an albumin-binding peptide [134]. This peptide ligand strongly interacts with albumin by noncovalent binding, which would facilitate easier dissociation of the peptide for release into the tumor. This PDC had a longer plasma elimination with a half-life of 29.4 ± 0.8 h compared with free DOX, which had a half-life of minutes. Meanwhile, the Dox in the tumor was found to be ≈ 120-fold greater for the PDC-treated mice compared to the free Dox group. However, considering the in vivo stability and prerelease of free drugs, the acid cleavable linkers are not widely explored as enzyme-cleavable linkers.

Succinic acid (SA) is another pH-cleavable linker moiety that has been successfully used for PDC construction [137]. This PDC was established by conjugating a hydrophilic cell-penetrating peptide (CPP = c[RGDKLAK]) with paclitaxel via ester linkage with succinic acid. This PDC demonstrated excellent solubility in water, proper drug releases, and prominent tumor-growth inhibitory effects in vivo. This study provides a new approach to treating glioblastoma.

Reducible disulfide linkers

Glutathione (GSH) is found in a fourfold higher concentration in the tumor environment than in normal cells, which had the advantage of a controlled release of the payload. Liang et al. designed a PDC with a disulfide linker between Dox and cyclic RGD peptide, and studied their drug release properties [38]. They found Dox in PDCs was released slowly over with 39.2% drug released at 48 h, but the release rate was dramatically increased in the presence of DTT (50 mM) to almost completion (100%) in 48 h, indicating the selective drug release in reductive environment. BicycleTx Limited (Cambridge, UK) screened and proved that a bicyclic PDC containing EphA2 peptide ligand, a disulfide linker, and cytotoxic mertansine (DM1), had potent in vivo anti-cancer effect, fast renal elimination and low accumulation in liver. Which demonstrates great potential for clinical cancer therapy [80]. However, the disulfide linker could sometimes influence the cell toxicity of the free drug. Li et al. designed a reduction-responsive PDC where a peptide BP9a analogue (CAHLHNRS) was linked to Dox through disulfide linker [138]. This PDC showed cell-selective toxicity towards HepG2 liver cancer cells, which are highly expressing transferrin receptors (TfR), but low potency compared to free Dox. To improve the drug efficacy, the combination with other linkers might be a good direction, like a disulfide linker in combination with hydrazone successfully used in two of the FDA approved ADCs.

Noncleavable linkers

Noncleavable linkers are not activated through external stimuli, which present increased stability in circulation, such as succinimidyl thioether, oxime, and triazole [139,140,141]. The cleavage of PDCs begins at the most labile group, most probably peptide being metabolized, resulting in different forms of drugs, including free drugs, drugs with a portion of the linker, or drugs with linker and amino acid(s). Gabor Mezo et al. designed and screened an oxime-linked daunorubicin (Dau) − GnRH-III conjugate with the incorporation of unnatural and D-amino acids in the homing peptides [142]. These PDCs exhibited high binding affinity for the GnRH receptors and excellent stability in 90% human serum for at least 24 h. This oxime linker was found to be highly stable to chemical or enzymatic degradation, and the conjugate got cleaved at the peptide portion and released the drug in tumor cells. Meanwhile, this PDC had good and selective tumor cell penetration and anti-tumor effect on MCF-7 and HT-29 cells. Randelovic et al. continued this study and revealed that this conjugate had a significant tumor volume reduction as well as metastases inhibition effect with less toxic side effects compared to the free drug in colorectal cancer or breast cancer-bearing mouse models [143]. However, it is not always lucky enough to construct PDCs with good stability, cellular selectivity, and potent efficiency. Feni et al. utilized oxime to attach cytotoxic daunorubicin to a cell penetration peptide, and additional chemistries like triazole and amide were utilized to put an RGD on this PDC [143]. This PDC showed no selectivity toward cells with higher expression of integrin αvβ3 (U87) and much less toxic than the free daunorubicin. This proper peptide ligand or chemistry used for the PDC design greatly influenced the efficacy and cell selectivity.

The non-cleavable triazole formed by the alkyne − azide reaction provides stability to the PDC structure, and other bonds in the linker region likely facilitate the release of the drug from the PDC after cellular uptake. Zheng et al. explored and screened PDCs for cancer photodynamic therapy (PDT) and found the distances between the phthalocyanine and the peptide impacted the photocytotoxicity of these PDCs significantly [141]. They found one conjugate had selective toxicity towards HT29 cells compared to MCF-7. The in vivo study revealed that this PDC had selective accumulation at the tumor site and led to a reduction of tumor growth by approximately 75%. This case mentioned that both the linker stability and length might influence the drug behavior.

Payloads

Despite the potent ability for tumor killing, toxin drugs usually can not satisfy clinical demands, such as poor PK properties and target tissue selectivity. The drug conjugation with peptides allows for specific targeted therapy, consequently resulting in the enhancement of several properties, including the therapeutic window and safer therapeutic dosage. There are several requirements for cytotoxic drugs to use in PDCs, including a clear mechanism of cellular action, a small molecular weight, high cytotoxicity, and retained anti-tumor activity after chemical coupling to the peptides [10]. The advantage of conjugating traditional cytotoxic drugs with peptides lies in the ability to modify their chemical properties, including solubility, selectivity, and half-life, thereby enhancing the efficacy of conventional therapies [144]. Besides chemotherapeutic drugs and radiotracers [145], other different types of drugs can also be designed to conjugate with peptides to improve their therapeutic properties, including small targeted drugs [108], anti-cancer peptides [55], protein drugs [146], nucleic acid drugs [147] and gas molecules [148, 149] et al. These different kinds of payloads enriched the application of peptide-based therapeutics and promoted the possibility of clinical transformation.

The chemotherapeutic drugs are always the first-line drugs for cancer patients, such as DOX, PTX, CPT, and Pt, which are the most commonly used chemo-agents in clinical practices. However, these drugs usually suffer from poor solubility, short circulation half-lives, potential development of drug resistance, and severe side effects, which significantly reduce their therapeutic outcomes and limit their clinical application. PDCs have a great potential to address these problems for targeted therapy, and toxic drugs such as DOX [150, 151], PTX [152,153,154,155], CPT [156], Pt [157] have been conjugated in PDCs for better clinical application.

In addition to chemo-agents, radionucleotides can also be used as therapeutic or imaging agents. Nowadays, many different radionucleotides have been used as payloads in PDCs for cancer diagnosis or treatment, including gallium-68 (68Ga), copper-64 (64Cu) and fluorine-18 (18 F),123I,99mTc et al. For example, 111In-DTPA-Octreotide (Octreoscan) was approved for the diagnosis of neuroendocrine tumors, and 177Lu-dotatate showed an increased period for progression-free survival for cancer patients and had greater potential as a therapeutic [15].

The gas therapy, including nitric oxide (NO), hydrogen sulfide (H2S), oxygen (O2), carbon monoxide (CO), and sulfur dioxide (SO2), is attracting great attention due to no drug-resistant, little side-effect, and no by-product, which showed great potential application in cancer treatment [10]. The lack of generators for the controlled release of gas limited their further application. Polypeptide-gas conjugates can achieve controlled release of gas and overcome multidrug resistance (MDR) and tumor recurrence [148, 149].

Small-molecule in targeted cancer therapy showed better tumor selectivity compared to chemotherapeutic drugs but also had limitations in the clinical applications, such as drug resistance, side effects, low efficiency, et al. [1]. Peptides can also be used as carriers to deliver this kind of drug to achieve high anti-cancer effects with low non-specific toxicity, such as peptide-HDACi conjugates by our group [107, 108]. In the future, peptide-targeted drug conjugation could be an alternative to overcome the challenges for clinical application.

Anti-cancer peptides can also be used as payloads for PDC construction, such as the most widely used lytic peptide [158] or pro-apoptotic peptide KLA [159, 160]. Like chemo-agents, this peptide can be conjugated with tumor-targeting peptide ligands to reduce the non-specific toxicity or conjugated with cell penetration peptides to increase their anti-cancer effects. We previously developed a VEGFR targeting peptide conjugate (QR-KLU), which showed good in vivo anti-tumor effect than traditional drug DOX through TACE in VX2 rabbit tumor model, and efficiently inhibited angiogenesis in tumor tissues with good safety, shown in Fig. 5 [55]. This work broadened the application of PDCs and may provide an alternative option for clinical HCC therapy via TACE combination.

Biomacromolecules such as protein drugs or nucleic acid drugs are novel and promising drug candidates for various refractoriness diseases. The main challenges of these drugs are poor cell penetration and tumor target ability. For example, NGR-human tumor necrosis factor (TNF) was designed to improve tumor targets and decrease their non-specific toxicity [52]. Small interfering RNA (siRNA) is regarded as a promising tool for cancer therapy because of its wide applicability to various cancer-related genes. Peptide-RNA conjugates could efficiently deliver RNA drugs to cancer cells to induce cell apoptosis [161]. However, due to the poor pharmaceutical properties of both peptide and RNA, this kind of conjugate is not suitable for clinical application. Nowadays, peptide and RNA are both encapsulated into various nanoparticles for targeted cancer therapy [162].

The pharmacokinetics of PDCs

A small molecule is considered “drug-like” and orally bioavailable if it adheres to Lipinski’s Rule of 5 (Ro5), which stipulates specific criteria for molecular weight, hydrogen bond donors, hydrogen bond acceptors, and log P [163, 164]. While peptides often do not meet these criteria, numerous marketed and clinical-stage peptide drugs demonstrate that Ro5 is not a definitive barrier to drug development. Understanding the ADME (Absorption, Distribution, Metabolism, Excretion) properties of peptides is crucial for designing effective PDCs for cancer therapy. The in vivo behavior of peptides significantly influences the pharmacokinetics (PK) of PDCs, ultimately impacting their therapeutic efficacy. However, the complex nature of ADME processes remains a subject of ongoing research. In this section, we will provide a concise overview of peptide administration, biodistribution, metabolism, and clearance based on current scientific knowledge. Our goal is to offer insights into the design of more effective PDC drugs for clinical application.

Absorption

Similar to ADCs, PDCs typically require parenteral administration to achieve optimal bioavailability and therapeutic efficacy. However, injections can be painful and may induce allergic reactions or serious side effects. Oral administration is the most convenient and preferred route for drug delivery. Nevertheless, the poor intestinal absorption of peptides due to enzymatic degradation and limited membrane permeability poses significant challenges for the oral delivery of PDCs.

Strategies developed for the oral delivery of peptide drugs can provide valuable insights for PDC oral delivery. Absorption enhancers, such as surfactants, bile acids, fatty acids, EDTA, and sodium salicylate, have been explored to improve the intestinal absorption of peptide drugs like insulin. The incomplete absorption of peptide drugs is often attributed to a combination of poor membrane permeability and metabolic degradation at the absorption site [165].

One major obstacle to the oral bioavailability of peptides is the extensive hydrolysis by digestive enzymes in the gastrointestinal tract. Protease inhibitors, such as aprotinin, trypsin, bacitracin, puromycin, and bestatin, can reduce peptide degradation at absorption sites, offering a potential strategy to improve peptide drug delivery [165]. Chemical modifications can enhance peptide oral absorption, including fatty acid conjugation [166], PEGylation [167, 168], cyclization, and the incorporation of unnatural amino acids [169,170,171].

CPPs can facilitate cell entry or membrane translocation, promising to increase peptide oral bioavailability [172]. Drug carrier systems, such as nanoparticles and hydrogels, can prolong gastric residence time and increase intimate contact with the intestinal epithelial layer, thereby improving drug absorption and oral bioavailability [173, 174]. However, despite these strategies, significant challenges remain in translating oral peptide drug delivery into clinical applications.

Biodistribution

The biodistribution of PDCs, like peptide drugs, is influenced by factors such as molecular weight, charge, protein binding, structure, and reliance on active transport. Conjugation to different molecules can alter the half-life, selective tissue distribution, or both of PDCs. Chemical modifications, which can enhance oral absorption, may also impact biodistribution.

Parisa et al. investigated the biodistribution and tumor localization of an ABD-Dox conjugate, where ABD is an albumin-binding peptide. This PDC exhibited approximately 4- and 2-fold greater tumor accumulation compared to free Dox and prodrug AlDox, respectively. Conversely, at 2 h post-administration, ABD-Dox accumulated at lower concentrations in the liver, spleen, kidney, and muscle compared to free Dox or AlDox [174]. These findings suggest that ABD-DOX may have reduced cardiotoxicity, hepatotoxicity, and nephrotoxicity, which are common side effects of free DOX. However, the mechanisms underlying PDC biodistribution are complex. Improving the enzymatic and chemical stability of peptides is a promising strategy to enhance targeted distribution.

Metabolism

PDCs, composed of peptides and small molecules, are susceptible to degradation by various proteases, including exopeptidases (such as aminopeptidases, carboxypeptidases, and dipeptidases) and endopeptidases (such as pepsin, trypsin, chymotrypsin, and elastase) [175]. These proteolytic enzymes play crucial roles in blood and cells, regulating peptide signaling by activating or deactivating proteins. The metabolism of small molecule drugs in PDCs is complex, as different molecules undergo diverse metabolic pathways catalyzed by various enzymes, including the cytochrome P450 family. While it is challenging to significantly alter the metabolic stability of small molecules significantly, stabilizing the peptide component can substantially impact the in vivo stability and efficacy of PDCs.

Renal clearance

While peptides often exhibit increased stability compared to small molecules, they can still be subject to rapid renal clearance. The in vivo clearance of peptides and PDCs is influenced by various factors, including molecular weight, charge, hydrophobicity, and enantiomeric purity [176]. It has been observed that peptides with a net negative charge tend to have longer half-lives compared to those with a net positive charge. The anionic nature of the glomerular membrane limits the filtration of negatively charged molecules into the urine [176]. However, negative charges can hinder cellular uptake, potentially affecting peptide function. Another strategy to prolong peptide half-life involves conjugation with larger molecules (> 450 kDa), such as PEGylation. This can increase lipophilicity and binding to plasma proteins like albumin, improving pharmacokinetic and pharmacodynamic properties [177]. For instance, PEGylation of the RGD-targeting HM-3 peptide significantly increased its half-life in rats. When this peptide was conjugated with a PEG linker attached at the N-terminus, the newly modified peptides had a 5.86-fold increase in half-life in male rat studies when compared to the unmodified HM-3 peptide [178]. The successful examples of prolonging peptide half-life in vivo are the remodeling of the insulin or GLP-1 analogs. In 2009, Liraglutide, a near analog of human GLP-1, had a fatty acid chain with a spacer joined to the main peptide backbone for binding to albumin. Liraglutide represented a significant improvement of GLP-1 with an extended IV half-life of 8–10 h and once-daily administration [179].

The recent advances of PDCs in clinical application

PDCs encompass a diverse range of conjugates utilizing various peptide types, including linear peptides with strong target affinity or cell penetration capabilities, cyclic or bicyclic peptides identified through phage display with enhanced stability and affinity, and peptide dendrimers or self-assembling peptides with favorable delivery properties. All three types of PDCs have entered clinical trials, demonstrating significant potential for clinical applications (Table 4).

Table 4 Recent advances in PDC clinical applications

Radiotracer peptide drug conjugates in clinical trails

Several radiolabeled PDCs have been approved for disease diagnosis or therapy. 99mTc-EDDA/HYNIC-3Tyr-octreotide was approved in Poland in 2013 for the treatment of neuroendocrine tumors of bronchial and thymic origin [180, 181]. Lu-DOTATATE (Lutathera), approved in the USA in 2018, is the first peptide-drug conjugate for the treatment of somatostatin receptor-positive gastroenteropancreatic neuroendocrine tumors via intramuscular injection [182]. 68Ga-DOTATOC (approved in the EU and USA in 2016–2019) and 68Ga-DOTATATE (approved by the FDA in 2016) are approved for the diagnosis of neuroendocrine tumors. 68Ga-DOTATOC, for example, exhibits 80% tumor accumulation within 30 min with a target-to-non-target ratio of 100:1 for central nervous system lesions compared to normal brain tissue [183, 184]. The tumor-targeting peptides used in these approved PDCs are Octreotide (the sequence D-Phe-Cys-Phe-D-Trp- Lys-Thr-Cys-Thr(ol) (fCFwKTCT-ol; 2–7 disulfide bond)) and its derivatives with good affinity to SST2 receptor subtype. Numerous other radiolabeled peptide-drug conjugates are currently undergoing clinical trials but will not be discussed further in this paper.

AEZS-108 (zoptarelin doxorubicin)

AEZS-108 (zoptarelin doxorubicin) is a cytotoxic hybrid molecule comprising doxorubicin covalently linked to a LHRH analog [71]. This design selectively targets doxorubicin to tumor cells expressing LHRH receptors. AEZS-108 has been evaluated in a Phase II trial in men with metastatic castrate-resistant prostate cancer who had progressed after taxane-based chemotherapy [71, 185]. Additionally, it is being tested in a Phase III trial in women with endometrial cancer resistant to platinum/taxane-based chemotherapy [186]. Preliminary data from the Phase II trial in men with metastatic castrate-resistant prostate cancer demonstrated activity in a challenging patient population and a favorable safety profile. However, in a large Phase III trial in advanced endometrial cancer, AEZS-108 failed to improve median overall survival or progression-free survival compared to standard doxorubicin therapy despite promising results in earlier Phase II studies in castration-resistant prostate cancer.

ANG1005 (Angiopep-2)

ANG1005, a novel PDC, consists of three paclitaxel molecules covalently linked to Angiopep-2. This design facilitates crossing the blood-brain and blood-cerebrospinal barriers and penetrating malignant cells via the LRP1 transport system [77]. A first-in-human study evaluated the safety, tolerability, pharmacokinetics, and efficacy of ANG1005 in patients with advanced solid tumors. The pharmacokinetics was dose linear, and the mean terminal-phase elimination half-life was 3.6 h. No evidence of accumulation was observed after repeat dosing [187]. No anti-GRN1005 antibodies were detected. The main dose-limiting toxicity was myelosuppression. Overall, ANG1005 demonstrated good tolerability and promising activity in heavily pretreated patients with advanced solid tumors, including those with brain metastases or prior failed taxane therapy. A subsequent open-label phase II study (NCT02048059) in adults with recurrent breast cancer brain metastases did not meet the primary endpoint of intracranial objective response rate (iORR) per independent radiology facility (IRF) evaluation. However, notable central nervous system (CNS) and systemic treatment effects were observed in all patients, including symptom improvement and prolonged overall survival compared to historical controls, particularly in the subset with leptomeningeal carcinomatosis (n = 28). Other registered clinical trials included ANG1005 in patients with recurrent high-grade glioma in the USA (NCT01967810) and ANG1005 alone or in combination with trastuzumab in breast cancer patients with brain metastases in the USA ( NCT01480583). However, no related results were posed on ClinicalTrials.gov.

CBX-12

CBX-12 is a clinically developed pH-sensitive peptide-exatecan conjugate (peptide sequence: ADDQNPWRAYLDLLFPDTLLLDLLW) that selectively targets cancer cells due to the acidic tumor microenvironment [188]. In vitro, plasma stability assays demonstrated robust linker stability and minimal warhead release in mouse, rat, dog, and human plasma samples at 24 h. CBX-12 effectively delivers high doses of exatecan directly to tumors, resulting in potent tumor cell killing across multiple xenograft models, including HCT116, MDA-MB-231, JIMT-1, and MKN45, with minimal to no bone marrow or life-threatening gastrointestinal toxicity.

The structure of CBX-12 reveals that it adopts the identical amino acid sequence as Variant 3 [189]. In vitro plasma stability tests conducted in mice, rats, dogs, and humans demonstrated excellent stability for CBX-12. No detectable toxin release was observed within 8 h, and only minimal toxin release was detected at 24 h, confirming its stability in blood circulation. Furthermore, in xenograft animal models, CBX-12 once again demonstrated both blood circulation stability and selectivity for tumor cells.

Most notably, compared to standalone Exatecan (DX-8951, Iritotecan), the peptide-conjugated CBX-12 exhibited significantly stronger tumor inhibitory effects across various tumor models [188, 190]. In treatments using unconjugated Exatecan, model animals consistently showed significant weight loss or fluctuations. In contrast, CBX-12 had a minimal impact on body weight, highlighting its tolerability advantage. Moreover, unconjugated Exatecan caused damage to multiple organs in the animals, including significant gastrointestinal (GI) toxicity observable under microscopy. In comparison, animals treated with CBX-12 showed no detectable GI toxicity.

A first-in-human, Phase 1/2 open-label, multicenter, dose-escalation study is currently recruiting subjects with advanced or metastatic refractory solid tumors to evaluate the safety, pharmacokinetics, and biomarker profile of CBX-12 (NCT04902872). We anticipate promising therapeutic efficacy from this clinical trial.

TH1902

TH1902 is a docetaxel-peptide conjugate designed to selectively target cancer cells overexpressing the sortilin (SORT1) receptor [191]. SORT1, also known as neurotensin receptor 3, is a membrane-bound receptor belonging to the VPS10P receptor family [192]. It plays diverse roles in intracellular transport and sorting of various ligands, including neurotensin, granulin precursor, and apolipoprotein E. On the other hand, SORT1 upregulation has been observed in several types of human cancers, such as breast cancer, ovarian cancer, pancreatic cancer, melanoma, and pituitary adenomas [193, 194]. Notably, SORT1 is expressed in 59% of TNBC cases, making it a potential therapeutic target for TNBC. In vitro studies on TNBC-derived MDA-MB-231 cells demonstrated that TH1902 exhibited potent anti-proliferative and anti-migratory activities, inducing faster and more potent apoptotic cell death compared to unconjugated docetaxel [191]. In vivo, studies using MDA-MB-231 and HCC-70 murine xenograft models revealed that both intraperitoneal and intravenous administration of TH1902 led to greater tumor regression than docetaxel without inducing neutropenia. These results highlight the high in vivo efficacy and safety of TH1902. Additionally, studies demonstrated that weekly administration of TH1902 was better tolerated than equivalent doses of docetaxel and effectively inhibited tumor growth in ovarian and endometrial xenograft models. As a single agent, TH1902 exhibited superior in vivo efficacy compared to both unconjugated taxanes and carboplatin against SORT1-positive ovarian and endometrial cancers. Currently, an open-label, first-in-human study of TH1902 in solid tumors is active but not currently recruiting (NCT04706962). In early 2021, the FDA granted TH1902 Fast Track designation for monotherapy in treating sortilin-positive recurrent advanced solid tumors that are resistant to standard therapies.

In TH1902, the peptide molecule specifically targets the SORT1 protein expressed in multiple cancers. A cleavable linker connects the peptide to the toxic payload, docetaxel, enabling targeted therapy. In terms of its mechanism of action, TH1902 applies the “lock-and-key theory” to conjugated drugs by competitively binding to the target SORT1.

Another similarity between PDCs and ADCs lies in their ability to achieve precise payload release, minimizing exposure to normal tissues and enhancing both tolerability and efficacy. In a sense, conjugated drugs function as a type of drug delivery technology, and preclinical data for TH1902 support this characteristic.

TH1902 is constructed with a 2:1 ratio of docetaxel to a peptide molecule connected via a linker (succinic acid). It enters cells through a sortilin-dependent mechanism, effectively bypassing the P-gp efflux pump (MDR1), thereby increasing intracellular concentrations. This results in improved tumor migration inhibition and growth suppression.

EP-100

EP-100 is a novel anti-cancer PDC composed of a natural LHRH ligand linked to a cationic membrane-disrupting peptide [195]. EP-100 interacts with negatively charged tumor cell membranes, inducing cell death through rapid membrane lysis. Preclinical studies have demonstrated the anti-tumor efficacy of EP-100 against various human cancer cell lines that overexpress GnRH-R, both alone and in combination with paclitaxel. A Phase 1 clinical trial (NCT00949559) evaluated EP-100 in patients with diverse solid tumors, including breast, ovarian, endometrial, pancreatic, prostate, colon, and non-Hodgkin lymphoma. The drug was found to be safe and well-tolerated [196]. In 2021, a multicenter, open-label, randomized Phase II trial (NCT01485848) compared paclitaxel plus EP-100 to paclitaxel alone in patients with refractory or recurrent ovarian cancer. While the overall response rate was similar between the two groups, a subset of patients with liver metastases appeared to benefit from the combination therapy. Importantly, the addition of EP-100 did not significantly increase the adverse event profile of paclitaxel and was well-tolerated [195].

G-202

Mipsagargin (G-202) is a novel, targeted prodrug based on thapsigargin, a potent inhibitor of the sarcoplasmic/endoplasmic reticulum calcium ATPase (SERCA) pump. Mipsagargin is designed to be activated by prostate-specific membrane antigen (PSMA)-mediated cleavage of an inert masking peptide, leading to the release of active thapsigargin within tumor cells and tumor vasculature [57]. While thapsigargin itself is poorly water-soluble and lacks a therapeutic index, the peptide conjugation improves its water solubility, enabling systemic delivery and selective release within PSMA-expressing tumors. A Phase I clinical trial (NCT01056029) demonstrated acceptable tolerability and favorable pharmacokinetics in patients with solid tumors [56]. A subsequent Phase II study (NCT01777594) evaluated Mipsagargin in advanced hepatocellular carcinoma patients who had progressed on or were intolerant to sorafenib. The drug was well-tolerated and showed promising disease stabilization in this patient population. However, a larger clinical trial to further characterize Mipsagargin’s activity in advanced HCC was unfortunately canceled due to company reorganization.

Additionally, a Phase II study (NCT02067156) to evaluate the efficacy, safety, and CNS exposure of Mipsagargin in patients with recurrent or progressive glioblastoma was completed in 2017, but results have not been publicly disclosed. Another Phase II study (NCT02381236) was also completed to assess the impact of Mipsagargin on prostate perfusion and volume, but the results remain undisclosed.

Bycycic peptide drug conjugates

Bicyclic toxin peptides (BTPs) are a promising class of PDCs composed of 9–20 amino acids with three cysteine residues that form disulfide bonds, creating a rigid cyclic structure. These disulfide bonds link the peptide to small molecule toxins, enhancing stability and target specificity. In 2021, Bicycle Therapeutics announced three investigational BTPs (BTCs) for Phase II/III clinical trials: (1) BT1718, targets membrane type I matrix metalloproteinase (MT1-MMP) to release the cytotoxic payload DM1 [75]; (2) BT5528, targets EphA2, a transmembrane tyrosine kinase receptor overexpressed in various cancers, including breast, colon, uroepithelial, lung, cervical, and ovarian cancers [197]; (3) BT8009, targets nectin-4, a type I membrane protein overexpressed in many tumors, including uroepithelial, breast, pancreatic, and triple-negative breast cancers [198]. A Phase I/IIa trial of BT1718 in patients with advanced solid tumors (NCT03486730) was initiated with the aim to study their pharmacokinetics and find the maximum dose of BT1718 and their potential side effects. The results revealed that BT1718 is well tolerated, and the pharmacokinetics are consistent with the proposed preclinical mechanism of tumor-targeted toxin delivery [199]. A phase I/II study of BT5528 in patients with advanced malignancies associated with EphA2 expression (NCT04180371) was carried out to study the drug safety, pharmacokinetics, and preliminary clinical activity. The results revealed that BT5528 was well tolerated with controllable side effects. They demonstrated favorable and preliminary anti-tumor activity, particularly in urothelial cancer. The pharmacokinetics are generally dose-proportional, with a short half-life (0.4–0.7 h) [200]. These results support further development of BT5528 as monotherapy and in combination with nivolumab and potentially other agents in select tumor types [201]. Two clinical studies of BT8009 have been recruited in patients with locally advanced or metastatic urothelial cancer(NCT06225596) and patients with Nectin-4 expressing advanced malignancies(NCT04561362). Both of these two trials will assess the safety and tolerability of BT8009 alone and in combination with pembrolizumab in patients with select advanced solid tumors. However, the results have not been published. Overall, the promising anti-tumor activity in preclinical and early clinical studies highlights the potential of BTPs.

As we described above, PDCs have made some progress in certain clinical trials. However, most of them are in phase I/II clinical studies, and the clinical efficacy and safety have not been largely investigated. The most promising PDCs are radionuclide-peptide conjugates, which exhibited good tumor targeting and therapeutic efficacy, such as the approved Lutathera for the treatment of patients with somatostatin receptor (SSTR)-positive gastroenteropancreatic neuroendocrine tumors and the approved 177Lu-PSMA-617 (Pluvicto) for the treatment of adult patients with prostate-specific membrane antigen (PSMA)-positive metastatic castration-resistant prostate cancer (mCRPC). The success of these two approved PDCs accelerated the development of radionuclide-peptide conjugates for the treatment of various malignant cancers. Pepaxto (Melflufen) is another approved PDC for the treatment of heavily pretreated relapsed and refractory multiple myeloma. However, this drug was withdrawn from the market due to the unsatisfied clinical outcomes in phase III clinical trials. As for other PDCs tested in clinical trials, the safety has been widely proven, but the efficacy is not convincing enough to get approval. Among these PDCs, ANG1005 (Angiopep-2) showed potential for cancer patients due to the good clinical results that notable central nervous system (CNS) and systemic treatment effects were observed in all patients treated with ANG1005 in phase II clinical trial, including symptom improvement and prolonged overall survival with good tolerability in heavily pretreated patients with advanced solid tumors with brain metastases or prior failed taxane therapy. Although the results of ANG1005 in phase III trials have not been revealed, this kind of drug modality provided a new approach or direction for refractory metastatic tumors. The bicyclic peptide drug conjugates also demonstrated great potential for cancer therapy due to the controlled side effects, good pharmacokinetics, and therapeutic activity.

Current challenges and future directions of PDCs

Current challenges of PDCs

In order to achieve their full therapeutic potential, PDC drugs must overcome enzymatic degradation in serum and effectively penetrate target tissues to release their payloads. Two primary strategies can be employed to enhance stability and cell penetration: active formulations and covalent modifications. Active formulations, such as polypeptides, nanoparticles, liposomal formulations, polymers, dendrimers, quantum dots, and polymeric micelles, have expanded the clinical applications of PDCs [202]. For instance, the FDA-approved Sandostatin LAR encapsulates octreotide within a glucose-poly(lactide-co-glycolide) (Glu-PLGA) polymer, extending its release profile. Polypeptide carriers like PEGylated poly(aspartic acid) (PAsp) and PEGylated poly(l-glutamic acid) (PLG) can prolong blood circulation time, enhance therapeutic efficacy, and reduce systemic toxicity of DOX [203]. Opaxio (CT-2103), a PLG-doxorubicin conjugate, demonstrates improved stability, comparable efficacy to free doxorubicin, and reduced adverse effects [204]. The conjugation of short hydrophilic peptides with hydrophobic drugs can induce self-assembly into supramolecular nanostructures, such as nanofiber hydrogels [205]. These structures can significantly improve drug delivery and efficacy.

Covalent modifications, including peptide cyclization, incorporation of unnatural amino acids or N-methylation, and conjugation with macromolecules, can enhance PDC stability and function [10]. The conformational constraints enforced by cyclization can render cyclic peptides highly resistant to proteolysis by endogenous proteases compared to their linear counterparts [206]. Various cyclization methods, such as disulfide bonds, lactam bridges, and stapling, have been explored [207]. For example, N-terminal crosslinked aspartic acid (TD) stapling offers a simple and cost-effective approach to stabilize peptides and improve their selectivity [107]. Besides improving peptide binding affinity and cellular uptake, peptide cyclization can enhance binding affinity and cellular uptake, as well as tissue penetration. Pentelute et al. demonstrated that perfluoroalkyl-cysteine SNAr chemistry could be used to create macrocyclic peptides with improved blood-brain barrier penetration. These macrocyclic peptides exhibited increased brain parenchyma penetration following intravenous administration in mice, suggesting potential applications for PDCs and peptide drugs in brain-related diseases [208]. Bicycles are another strategy involving the cross-linking of three cysteine residues to a trifunctional linker, forming a rigid cyclic structure. This approach can enhance the stability, selectivity, and potency of peptide-based drugs. The modern technologies for bioactive cyclic peptide screening accelerate the development of optimal drug candidates for binding targets with high affinity and specificity, such as phase display [40], one-bead-one-compound (OBOC) method [209], mRNA display [210], split-intein circular ligation of peptides and proteins (SICLOPPS) et al. [211].

D-amino acids can reduce susceptibility to proteolytic enzymes, increasing peptide stability. Octreotide, a peptide hormone containing two D-amino acids, exhibits a significantly longer half-life (1.5 h) compared to the unmodified peptide somatostatin (a few minutes) [181]. N-methylation can enhance peptide stability by increasing steric hindrance and altering peptide conformation. Cyclosporin, a hepta-N-methylated cyclic undecapeptide, is a well-known example of a highly stable and orally bioavailable peptide drug [212]. PEGylation is a common strategy to improve peptide pharmacokinetic properties [213,214,215]. PEGylation can increase molecular weight, reduce renal clearance, and prolong circulation time. For instance, PEGylation of the HM-3 peptide significantly extended its half-life [178]. Lipid conjugation is another approach to enhance peptide stability and pharmacokinetic properties. Semaglutide, a GLP-1 analog, is conjugated to a fatty acid chain, resulting in a significantly longer half-life and enabling once-weekly administration [179]. Thus, the chemical modification of peptides can not only improve PDC’s pharmacokinetic and pharmacodynamic properties for absorption but also have a pronounced effect on cell membrane permeability and target affinity to reach intracellular targets.

Besides the peptide stability, effective drug release is crucial for therapeutic efficacy, which depends on linker structures to break off in specific environments (temperature, pH, enzymes, redox, etc.). Despite different kinds of linkers being designed, toxic drugs sometimes can not be released as a prototype drug or even not be released at targeted tissues, which demonstrates weaker effects than prototype drugs. Alas et al. reviewed different linkers used in PDCs between the peptide and the linker and between the drugs and the linkers for cancer therapy [11]. The ideal linkers conjugate to homing peptides should not affect the binding affinity for its receptor, and the linkers attached to drugs can be cleaved to release the unmodified drug at the target site. Thus, linker chemistry plays a major role in the success of a PDC and in improving efficacy.

In summary, the chemical structure of both peptides and linkers used in the design and development of PDCs play key roles in their clinical applications.

Future directions of PDCs

Bicycle-toxin conjugates (BTCs)

Contrary to the common belief that cyclic peptides are inherently more stable than linear peptides, they can still be susceptible to rapid in vivo degradation [216]. Bicyclic peptides, with their additional cyclic constraint, offer enhanced proteolytic stability. BTCs have emerged as promising drug delivery systems (Fig. 7). These conjugates typically consist of a 9–20 amino acid bicyclic peptide linked to a small molecule toxin [217]. The rigid, cyclic structure of the peptide ensures the toxin’s conformation remains intact, optimizing its activity [198]. BTCs offer several advantages over ADCs, including deeper tumor penetration, rapid extravasation, and slower renal clearance. Bicycle Therapeutics is at the forefront of BTC development, with several compounds, including BT1718, BT5528, and BT8009, currently in clinical trials. These BTCs target specific tumor markers and have shown promising anti-tumor activity.

Advanced screening technologies, such as phage display, mRNA display, split intein circular ligation of peptides, and in silico screening, have revolutionized cyclic peptide drug discovery [218]. These technologies enable the rapid generation and screening of diverse cyclic peptide libraries, facilitating the identification of potent drug candidates. Structure-based drug design is another powerful approach to developing bicyclic peptides. By understanding the molecular interactions between a target protein and a peptide ligand, researchers can design bicyclic peptides with high affinity and specificity. For example, Grossmann et al. designed a library of bicyclic β-sheet mimetics that target β-catenin, a key player in Wnt signaling [219]. These peptides could serve as valuable building blocks for the development of potent BTCs.

Fig. 7
figure 7

Three different kinds of PDCs in the future development

Peptide dendrimer drug conjugates

Dendrimers are characterized by their highly branched, multilayered architecture, consisting of a core, multiple generations of branching, and a functionalized outer shell (Fig. 7) [220, 221]. Unlike linear peptides and polymers, dendrimers possess unique properties, including thermal stability, viscosity, and encapsulation capabilities [222]. Peptide dendrimers can be classified into two main types: covalent and noncovalent. Covalent peptide dendrimers are constructed using natural or unnatural amino acids, while noncovalent peptide dendrimers rely on noncovalent interactions to form their structure [223]. Peptide dendrimers have emerged as promising drug delivery vehicles. They can be used to deliver drugs through covalent conjugation or noncovalent encapsulation. For instance, Gu and colleagues demonstrated the successful use of peptide dendrimers to selectively deliver doxorubicin to cancer cells [224]. The incorporation of polyethylene glycol (PEG) into dendrimer nanoparticles can further enhance biocompatibility, reduce toxicity, and improve drug delivery efficiency.

Self-assembling peptide drug conjugates

Self-assembling peptide-based nanostructures offer significant advantages, including sustained drug release, precise targeting, overcoming drug resistance, and reducing off-target toxicity(Fig. 7) [122, 225]. As discussed in the Part 3.1.3 “Self-assembling peptide” of this review, various PDCs can self-assemble into nanostructures to enhance drug delivery. In order to address the challenges associated with clinical translation, innovative approaches are being developed to optimize the design and formulation of self-assembling peptide-based drug delivery systems. For example, the self-assembly of CXCR4 antagonist peptide-docetaxel conjugates, could self-assemble to nanoparticles, targeting CXCR4-upregulated metastatic tumor cells and enhancing the DTX efficacy. This self-assembly achieved promising efficacy on inhibiting both bone-specific metastasis and lung metastasis of triple-negative breast cancer [226]. These advancements hold the potential to revolutionize the treatment of various diseases.

Conclusion and discussion

PDCs hold immense potential as next-generation targeted therapeutics, offering advantages over ADCs in terms of synthesis, immunogenicity, and potency. However, challenges such as poor stability, low bioavailability, and lengthy development timelines hinder their clinical translation. PDCs and ADCs share fundamental similarities, and the clinical success of ADCs can inform the development of PDCs. Key challenges in ADC development, including on-target toxicity, linker instability, immune-related toxicity, receptor-mediated toxicity, and drug resistance, can guide the optimization of PDC design. In order to minimize off-target toxicity, ideal targets for PDCs should be selectively expressed on tumor cells. Recent advancements in gene sequencing and antigen discovery have enabled the identification of neoantigens and tumor-specific biomarkers. Additionally, targeting stromal cells, which share common markers across different tumor types, offers a promising strategy for broad-spectrum cancer therapy. Machine learning-based biology analysis can further accelerate the discovery of novel therapeutic targets.

Besides targeting the tumor-specific proteins to improve tumor selective toxicity, various strategies can be employed to enhance peptide stability and extend their circulation time: (1) Peptide Cyclization: Improves stability and resistance to proteolytic degradation. (2) Unnatural Amino Acid Incorporation: Enhances stability and alters peptide properties. (3) Macromolecular Conjugation: Increases molecular weight, reduces renal clearance, and prolongs circulation time (e.g., PEGylation). The choice of payload and linker is crucial for PDC efficacy and safety. Potent cytotoxic agents, such as small molecule drugs and toxins, can be conjugated to peptides. Additionally, emerging modalities like PROTACs and oligonucleotides offer exciting opportunities for PDC development. PDCs can be designed to address drug resistance mechanisms, such as increased drug efflux, altered target expression, and activated downstream signaling pathways. For example, targeting HSP90α, a protein involved in multiple drug resistance pathways, can sensitize tumor cells to therapy.

PDCs are currently limited to intravenous administration and cannot be delivered orally, posing a significant challenge for patients who are unable to attend regular follow-up visits. Recent studies have reported a novel method for achieving oral delivery of bioactive peptides using anionic nanoparticles [227]. While the use of nanoparticles is an attractive strategy to address the challenges of low oral bioavailability and stability, further testing is needed to assess the feasibility of this approach in future clinical trials. Additionally, alternative strategies, such as acid-resistant coatings, gut enzyme inhibitors, and mucus-penetrating peptides, have been proposed to improve the oral availability of PDCs [26]. These advancements promise to expand the accessibility and ease of administration of PDC therapies.

Meanwhile, the industrialization challenges of ADCs such as production costs and quality control also promote the development of PDCs. ADC production is expensive, time-consuming, and requires specialized facilities. Scaling up ADC production while maintaining consistency is difficult and costly. PDCs have lower production costs as peptides are chemically synthesized, relatively inexpensive process compared to antibody production. The conjugation of peptides, linkers, and payloads is simpler and more cost-effective than ADC conjugation [18]. ADCs are inherently heterogeneous due to variations in Drug-to-Antibody Ratio (DAR), conjugation sites, and antibody glycosylation patterns [19]. This requires highly stringent quality control (QC). While, PDCs are more homogeneous than ADCs as peptides are smaller and easier to characterize [18]. QC for PDCs are less stringent relying on simpler techniques like HPLC and mass spectrometry.

In conclusion, while the field of PDC development is still in its early stages, significant progress has been made. By addressing challenges in stability, delivery, and efficacy, PDCs have the potential to revolutionize cancer therapy. As our understanding of cancer biology and drug delivery mechanisms deepens, we can expect to see increasingly sophisticated PDC designs that offer improved therapeutic outcomes.

Data availability

No datasets were generated or analysed during the current study.

References

  1. Zhong L, Li Y, Xiong L, Wang W, Wu M, Yuan T, Yang W, Tian C, Miao Z, Wang T, Yang S. Small molecules in targeted cancer therapy: advances, challenges, and future perspectives. Signal Transduct Target Ther. 2021;6(1):201.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Ghosh S, Lai JY. An insight into the dual role of MoS2-based nanocarriers in anticancer drug delivery and therapy. Acta Biomater. 2024;179:36–60.

    Article  CAS  PubMed  Google Scholar 

  3. Zhou L, Lu Y, Liu W, Wang S, Wang L, Zheng P, Zi G, Liu H, Liu W, Wei S. Drug conjugates for the treatment of lung cancer: from drug discovery to clinical practice. Exp Hematol Oncol. 2024;13(1):26.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Yang Y, Wang S, Ma P, Jiang Y, Cheng K, Yu Y, Jiang N, Miao H, Tang Q, Liu F, Zha Y, Li N. Drug conjugate-based anticancer therapy - Current status and perspectives. Cancer Lett. 2023;552:215969.

    Article  CAS  PubMed  Google Scholar 

  5. Fu Z, Li S, Han S, Shi C, Zhang Y. Antibody drug conjugate: the biological missile for targeted cancer therapy. Signal Transduct Target Ther. 2022;7(1):93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Beck A, Goetsch L, Dumontet C, Corvaia N. Strategies and challenges for the next generation of antibody-drug conjugates. Nat Rev Drug Discov. 2017;16(5):315–37.

    Article  CAS  PubMed  Google Scholar 

  7. Liu M, Li L, Jin D, Liu Y. Nanobody-A versatile tool for cancer diagnosis and therapeutics. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2021;13(4):e1697.

    Article  CAS  PubMed  Google Scholar 

  8. Yu X, Long Y, Chen B, Tong Y, Shan M, Jia X, Hu C, Liu M, Zhou J, Tang F, Lu H, Chen R, Xu P, Huang W, Ren J, Wan Y, Sun J, Li J, Jin G, Gong L. PD-L1/TLR7 dual-targeting nanobody-drug conjugate mediates potent tumor regression via elevating tumor immunogenicity in a host-expressed PD-L1 bias-dependent way. J Immunother Cancer 10(10) (2022).

  9. Nessler I, Khera E, Vance S, Kopp A, Qiu Q, Keating TA, Abu-Yousif AO, Sandal T, Legg J, Thompson L, Goodwin N, Thurber GM. Increased tumor penetration of Single-Domain Antibody-Drug conjugates improves in vivo efficacy in prostate Cancer models. Cancer Res. 2020;80(6):1268–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Cooper BM, Iegre J, DH OD, Olwegard Halvarsson M, Spring DR. Peptides as a platform for targeted therapeutics for cancer: peptide-drug conjugates (PDCs). Chem Soc Rev. 2021;50(3):1480–94.

    Article  CAS  PubMed  Google Scholar 

  11. Alas M, Saghaeidehkordi A, Kaur K. Peptide-Drug conjugates with different linkers for Cancer therapy. J Med Chem. 2021;64(1):216–32.

    Article  CAS  PubMed  Google Scholar 

  12. Sharma K, Sharma KK, Sharma A, Jain R. Peptide-based drug discovery: current status and recent advances. Drug Discov Today. 2023;28(2):103464.

    Article  CAS  PubMed  Google Scholar 

  13. Dean TT, Jelu-Reyes J, Allen AC, Moore TW. Peptide-Drug conjugates: an emerging direction for the next generation of peptide therapeutics. J Med Chem. 2024;67(3):1641–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Paiva TB, Paiva AC, Freer RJ, Stewart JM. Alkylating analogs of peptide hormones. 2. Synthesis and properties of p-(N,N-bis(2-chloroethyl)amino)phenylbutyryl derivatives of angiotensin II. J Med Chem. 1972;15(1):6–8.

    Article  CAS  PubMed  Google Scholar 

  15. Zhu YS, Tang K, Lv J. Peptide-drug conjugate-based novel molecular drug delivery system in cancer. Trends Pharmacol Sci. 2021;42(10):857–69.

    Article  CAS  PubMed  Google Scholar 

  16. Wu M, Huang W, Yang N, Liu Y. Learn from antibody-drug conjugates: consideration in the future construction of peptide-drug conjugates for cancer therapy. Exp Hematol Oncol. 2022;11(1):93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Heh E, Allen J, Ramirez F, Lovasz D, Fernandez L, Hogg T, Riva H, Holland N, Chacon J. Peptide drug conjugates and their role in Cancer therapy. Int J Mol Sci 24(1) (2023).

  18. Zhang B, Wang M, Sun L, Liu J, Yin L, Xia M, Zhang L, Liu X, Cheng Y. Recent advances in targeted Cancer therapy: are PDCs the next generation of ADCs? J Med Chem. 2024;67(14):11469–87.

    Article  CAS  PubMed  Google Scholar 

  19. Li M, Zhao X, Yu C, Wang L. Antibody-Drug conjugate overview: a State-of-the-art manufacturing process and control strategy. Pharm Res. 2024;41(3):419–40.

    Article  PubMed  Google Scholar 

  20. Dumontet C, Reichert JM, Senter PD, Lambert JM, Beck A. Antibody-drug conjugates come of age in oncology. Nat Rev Drug Discov. 2023;22(8):641–61.

    Article  CAS  PubMed  Google Scholar 

  21. Scott JK, Smith GP. Searching for peptide ligands with an epitope library. Science. 1990;249(4967):386–90.

    Article  CAS  PubMed  Google Scholar 

  22. Lam KS, Lebl M, Krchnak V. The One-Bead-One-Compound combinatorial library method. Chem Rev. 1997;97(2):411–48.

    Article  CAS  PubMed  Google Scholar 

  23. Josephson K, Ricardo A, Szostak JW. mRNA display: from basic principles to macrocycle drug discovery. Drug Discov Today. 2014;19(4):388–99.

    Article  CAS  PubMed  Google Scholar 

  24. Lennard KR, Tavassoli A. Peptides come round: using SICLOPPS libraries for early stage drug discovery. Chemistry. 2014;20(34):10608–14.

    Article  CAS  PubMed  Google Scholar 

  25. Lau YH, de Andrade P, Wu Y, Spring DR. Peptide stapling techniques based on different macrocyclisation chemistries. Chem Soc Rev. 2015;44(1):91–102.

    Article  CAS  PubMed  Google Scholar 

  26. Drucker DJ. Advances in oral peptide therapeutics. Nat Rev Drug Discov. 2020;19(4):277–89.

    Article  CAS  PubMed  Google Scholar 

  27. Werle M, Bernkop-Schnurch A. Strategies to improve plasma half life time of peptide and protein drugs. Amino Acids. 2006;30(4):351–67.

    Article  CAS  PubMed  Google Scholar 

  28. Lucas AT, Robinson R, Schorzman AN, Piscitelli JA, Razo JF, Zamboni WC. Pharmacologic considerations in the disposition of antibodies and Antibody-Drug conjugates in preclinical models and in patients. Antibodies (Basel) 8(1) (2019).

  29. Teicher BA, Chari RV. Antibody conjugate therapeutics: challenges and potential. Clin Cancer Res. 2011;17(20):6389–97.

    Article  CAS  PubMed  Google Scholar 

  30. Schwach J, Abdellatif M, Stengl A. More than Toxins-Current prospects in designing the next generation of antibody drug conjugates. Front Biosci (Landmark Ed). 2022;27(8):240.

    Article  CAS  PubMed  Google Scholar 

  31. Garcia-Alonso S, Ocana A, Pandiella A. Resistance to Antibody-Drug conjugates. Cancer Res. 2018;78(9):2159–65.

    Article  CAS  PubMed  Google Scholar 

  32. Saleh K, Khoury R, Khalife N, Chahine C, Ibrahim R, Tikriti Z, Le Cesne A. Mechanisms of action and resistance to anti-HER2 antibody-drug conjugates in breast cancer. Cancer Drug Resist. 2024;7:22.

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Chang HL, Schwettmann B, McArthur HL, Chan IS. Antibody-drug conjugates in breast cancer: overcoming resistance and boosting immune response. J Clin Invest 133(18) (2023).

  34. Takegawa N, Nonagase Y, Yonesaka K, Sakai K, Maenishi O, Ogitani Y, Tamura T, Nishio K, Nakagawa K, Tsurutani J. DS-8201a, a new HER2-targeting antibody-drug conjugate incorporating a novel DNA topoisomerase I inhibitor, overcomes HER2-positive gastric cancer T-DM1 resistance. Int J Cancer. 2017;141(8):1682–9.

    Article  CAS  PubMed  Google Scholar 

  35. Kovtun YV, Audette CA, Mayo MF, Jones GE, Doherty H, Maloney EK, Erickson HK, Sun X, Wilhelm S, Ab O, Lai KC, Widdison WC, Kellogg B, Johnson H, Pinkas J, Lutz RJ, Singh R, Goldmacher VS, Chari RV. Antibody-maytansinoid conjugates designed to bypass multidrug resistance. Cancer Res. 2010;70(6):2528–37.

    Article  CAS  PubMed  Google Scholar 

  36. Li JY, Perry SR, Muniz-Medina V, Wang X, Wetzel LK, Rebelatto MC, Hinrichs MJ, Bezabeh BZ, Fleming RL, Dimasi N, Feng H, Toader D, Yuan AQ, Xu L, Lin J, Gao C, Wu H, Dixit R, Osbourn JK, Coats SR. A biparatopic HER2-Targeting Antibody-Drug conjugate induces tumor regression in primary models refractory to or ineligible for HER2-Targeted therapy. Cancer Cell. 2016;29(1):117–29.

    Article  CAS  PubMed  Google Scholar 

  37. Wang Y, Cheetham AG, Angacian G, Su H, Xie L, Cui H. Peptide-drug conjugates as effective prodrug strategies for targeted delivery. Adv Drug Deliv Rev. 2017;110–111:112–26.

    Article  PubMed  Google Scholar 

  38. Liang Y, Li S, Wang X, Zhang Y, Sun Y, Wang Y, Wang X, He B, Dai W, Zhang H, Wang X, Zhang Q. A comparative study of the antitumor efficacy of peptide-doxorubicin conjugates with different linkers. J Control Release. 2018;275:129–41.

    Article  CAS  PubMed  Google Scholar 

  39. Gong L, Zhao H, Liu Y, Wu H, Liu C, Chang S, Chen L, Jin M, Wang Q, Gao Z, Huang W. Research advances in peptide–drug conjugates. Acta Pharm Sin B. 2023;13(9):3659–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Zhou X, Smith QR, Liu X. Brain penetrating peptides and peptide-drug conjugates to overcome the blood-brain barrier and target CNS diseases. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2021;13(4):e1695.

    Article  PubMed  Google Scholar 

  41. Sharma R, Kallur KG, Ryu JS, Parameswaran RV, Lindman H, Avril N, Gleeson FV, Lee JD, Lee KH, O’Doherty MJ, Groves AM, Miller MP, Somer EJ, Coombes CR, Aboagye EO. Multicenter reproducibility of 18F-Fluciclatide PET imaging in subjects with solid tumors. J Nucl Med. 2015;56(12):1855–61.

    Article  CAS  PubMed  Google Scholar 

  42. Provost C, Prignon A, Rozenblum-Beddok L, Bruyer Q, Dumont S, Merabtene F, Nataf V, Bouteiller C, Talbot JN. Comparison and evaluation of two RGD peptides labelled with (68)Ga or (18)F for PET imaging of angiogenesis in animal models of human glioblastoma or lung carcinoma. Oncotarget. 2018;9(27):19307–16.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Liu Z, Niu G, Wang F, Chen X. (68)Ga-labeled NOTA-RGD-BBN peptide for dual integrin and GRPR-targeted tumor imaging. Eur J Nucl Med Mol Imaging. 2009;36(9):1483–94.

    Article  CAS  PubMed  Google Scholar 

  44. Zhao L, Wen X, Xu W, Pang Y, Sun L, Wu X, Xu P, Zhang J, Guo Z, Lin Q, Chen X, Chen H. Clinical evaluation of (68)Ga-FAPI-RGD for imaging of fibroblast activation protein and integrin alpha(v)beta(3) in various Cancer types. J Nucl Med. 2023;64(8):1210–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Ryppa C, Mann-Steinberg H, Fichtner I, Weber H, Satchi-Fainaro R, Biniossek ML, Kratz F. In vitro and in vivo evaluation of doxorubicin conjugates with the divalent peptide E-[c(RGDfK)2] that targets integrin alphavbeta3. Bioconjug Chem. 2008;19(7):1414–22.

    Article  CAS  PubMed  Google Scholar 

  46. Singh T, Kim TW, Murthy ASN, Paul M, Sepay N, Jeong Kong H, Sung Ryu J, Rim Koo N, Yoon S, Song KH, Jun Baek M, Jeon S, Im J. Tumor-homing peptide iRGD-conjugate enhances tumor accumulation of camptothecin for colon cancer therapy. Eur J Med Chem. 2024;265:116050.

    Article  CAS  PubMed  Google Scholar 

  47. Chen X, Plasencia C, Hou Y, Neamati N. Synthesis and biological evaluation of dimeric RGD peptide-paclitaxel conjugate as a model for integrin-targeted drug delivery. J Med Chem. 2005;48(4):1098–106.

    Article  CAS  PubMed  Google Scholar 

  48. Gandioso A, Shaili E, Massaguer A, Artigas G, Gonzalez-Canto A, Woods JA, Sadler PJ, Marchan V. An integrin-targeted photoactivatable Pt(IV) complex as a selective anticancer pro-drug: synthesis and photoactivation studies. Chem Commun (Camb). 2015;51(44):9169–72.

    Article  CAS  PubMed  Google Scholar 

  49. Vats K, Satpati D, Sharma R, Kumar C, Sarma HD, Dash A. (99m) Tc-labeled NGR-chlorambucil conjugate, (99m) Tc-HYNIC-CLB-c(NGR) for targeted chemotherapy and molecular imaging, J Labelled Comp Radiopharm 60(9) (2017) 431–438.

  50. Tripodi AAP, Toth S, Enyedi KN, Schlosser G, Szakacs G, Mezo G. Development of novel Cyclic NGR peptide-daunomycin conjugates with dual targeting property. Beilstein J Org Chem. 2018;14:911–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Shokri B, Zarghi A, Shahhoseini S, Mohammadi R, Kobarfard F. Design, synthesis and biological evaluation of peptide-NSAID conjugates for targeted cancer therapy. Arch Pharm (Weinheim). 2019;352(8):e1800379.

    Article  PubMed  Google Scholar 

  52. Valentinis B, Porcellini S, Asperti C, Cota M, Zhou D, Di Matteo P, Garau G, Zucchelli C, Avanzi NR, Rizzardi GP, Degano M, Musco G, Traversari C. Mechanism of action of the tumor vessel targeting agent NGR-hTNF: role of both NGR peptide and hTNF in cell binding and signaling. Int J Mol Sci 20(18) (2019).

  53. Bieker R, Kessler T, Schwoppe C, Padro T, Persigehl T, Bremer C, Dreischaluck J, Kolkmeyer A, Heindel W, Mesters RM, Berdel WE. Infarction of tumor vessels by NGR-peptide-directed targeting of tissue factor: experimental results and first-in-man experience. Blood. 2009;113(20):5019–27.

    Article  CAS  PubMed  Google Scholar 

  54. Tao Y, Du M, Zhu M, Wang Y, Fei Y, Zhao YQ, Ma J, Fan R, Dai F, Chen J, Yin J, Fan B, Zeng G. Antitumor effect of Peptide-Camptothecin conjugate targeting CD133 protein. Bioconjug Chem. 2024;35(11):1859–69.

    Article  CAS  PubMed  Google Scholar 

  55. Wang D, Liu J, Li T, Wang Y, Liu X, Bai Y, Wang C, Ju S, Huang S, Yang C, Zhou C, Zhang Y, Xiong B. A VEGFR targeting peptide-drug conjugate (PDC) suppresses tumor angiogenesis in a TACE model for hepatocellular carcinoma therapy. Cell Death Discov. 2022;8(1):411.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Mahalingam D, Wilding G, Denmeade S, Sarantopoulas J, Cosgrove D, Cetnar J, Azad N, Bruce J, Kurman M, Allgood V.E., Carducci M. Mipsagargin, a novel thapsigargin-based PSMA-activated prodrug: results of a first-in-man phase I clinical trial in patients with refractory, advanced or metastatic solid tumours. Br J Cancer. 2016;114(9):986–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Mahalingam D, Peguero J, Cen P, Arora SP, Sarantopoulos J, Rowe J, Allgood V, Tubb B, Campos L. A phase II, multicenter, Single-Arm study of Mipsagargin (G-202) as a Second-Line therapy following Sorafenib for adult patients with progressive advanced hepatocellular carcinoma. Cancers (Basel) 11(6) (2019).

  58. Kim EH, Lee J, Kwak G, Jang H, Kim H, Cho H, Jang Y, Choi J, Chi SG, Kim K, Kwon IC, Yang Y, Kim SH. PDL1-binding peptide/anti-miRNA21 conjugate as a therapeutic modality for PD-L1(high) tumors and TAMs. J Control Release. 2022;345:62–74.

    Article  PubMed  Google Scholar 

  59. Moon Y, Shim MK, Choi J, Yang S, Kim J, Yun WS, Cho H, Park JY, Kim Y, Seong JK, Kim K. Anti-PD-L1 peptide-conjugated prodrug nanoparticles for targeted cancer immunotherapy combining PD-L1 Blockade with Immunogenic cell death. Theranostics. 2022;12(5):1999–2014.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Wang X, Wang Y, Hu J, Xu H. An antitumor peptide RS17-targeted CD47, design, synthesis, and antitumor activity. Cancer Med. 2021;10(6):2125–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Mullapudi SS, Rahmat JN, Mahendran R, Lim YK, Ong LT, Wong KY, Chiong E, Kang ET, Neoh KG. Tumor-targeting albumin nanoparticles as an efficacious drug delivery system and potential diagnostic tool in non-muscle-invasive bladder cancer therapy. Nanomedicine. 2022;46:102600.

    Article  CAS  PubMed  Google Scholar 

  62. Sun LC, Coy DH. Somatostatin receptor-targeted anti-cancer therapy. Curr Drug Deliv. 2011;8(1):2–10.

    Article  CAS  PubMed  Google Scholar 

  63. Kwekkeboom DJ, Kooij PP, Bakker WH, Macke HR, Krenning EP. Comparison of 111In-DOTA-Tyr3-octreotide and 111In-DTPA-octreotide in the same patients: biodistribution, kinetics, organ and tumor uptake. J Nucl Med. 1999;40(5):762–7.

    CAS  PubMed  Google Scholar 

  64. Qiao Z, Zhang J, Jin X, Huo L, Zhu Z, Xing H, Li F. 99mTc-HYNIC-TOC imaging in the evaluation of pancreatic masses which are potential neuroendocrine tumors. Clin Nucl Med. 2015;40(5):397–400.

    Article  PubMed  Google Scholar 

  65. Kowalski J, Henze M, Schuhmacher J, Macke HR, Hofmann M, Haberkorn U. Evaluation of positron emission tomography imaging using [68Ga]-DOTA-D Phe(1)-Tyr(3)-Octreotide in comparison to [111In]-DTPAOC SPECT. First results in patients with neuroendocrine tumors. Mol Imaging Biol. 2003;5(1):42–8.

    Article  PubMed  Google Scholar 

  66. Lelle M, Kaloyanova S, Freidel C, Theodoropoulou M, Musheev M, Niehrs C, Stalla G, Peneva K. Octreotide-Mediated Tumor-Targeted drug delivery via a cleavable Doxorubicin-Peptide conjugate. Mol Pharm. 2015;12(12):4290–300.

    Article  CAS  PubMed  Google Scholar 

  67. Chen X, Zhang XY, Shen Y, Fan LL, Ren ML, Wu YP. Synthetic Paclitaxel-octreotide conjugate reversing the resistance of A2780/Taxol to Paclitaxel in xenografted tumor in nude mice. Oncotarget. 2016;7(50):83451–61.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Su Z, Niu J, Xiao Y, Ping Q, Sun M, Huang A, You W, Sang X, Yuan D. Effect of octreotide-polyethylene glycol(100) monostearate modification on the pharmacokinetics and cellular uptake of nanostructured lipid carrier loaded with hydroxycamptothecine. Mol Pharm. 2011;8(5):1641–51.

    Article  CAS  PubMed  Google Scholar 

  69. Ghaly HSA, Varamini P. New drug delivery strategies targeting the GnRH receptor in breast and other cancers. Endocr Relat Cancer. 2021;28(11):R251–69.

    Article  CAS  PubMed  Google Scholar 

  70. Vankadara S, Ke Z, Wang S, Foo SY, Gunaratne J, Lee MA, Koh X, Chia CSB. Cytotoxic activity and cell specificity of a novel LHRH peptide drug conjugate, D-Cys6-LHRH Vedotin, against ovarian cancer cell lines. Chem Biol Drug Des. 2024;103(4):e14516.

    Article  CAS  PubMed  Google Scholar 

  71. Liu SV, Tsao-Wei DD, Xiong S, Groshen S, Dorff TB, Quinn DI, Tai YC, Engel J, Hawes D, Schally AV, Pinski JK. Phase I, dose-escalation study of the targeted cytotoxic LHRH analog AEZS-108 in patients with castration- and taxane-resistant prostate cancer. Clin Cancer Res. 2014;20(24):6277–83.

    Article  CAS  PubMed  Google Scholar 

  72. Emons G, Gorchev G, Sehouli J, Wimberger P, Stahle A, Hanker L, Hilpert F, Sindermann H, Grundker C, Harter P. Efficacy and safety of AEZS-108 (INN: Zoptarelin doxorubicin acetate) an LHRH agonist linked to doxorubicin in women with platinum refractory or resistant ovarian cancer expressing LHRH receptors: a multicenter phase II trial of the ago-study group (AGO GYN 5). Gynecol Oncol. 2014;133(3):427–32.

    Article  CAS  PubMed  Google Scholar 

  73. Argyros O, Karampelas T, Asvos X, Varela A, Sayyad N, Papakyriakou A, Davos CH, Tzakos AG, Fokas D, Tamvakopoulos C. Peptide-Drug conjugate GnRH-Sunitinib targets angiogenesis selectively at the site of action to inhibit tumor growth. Cancer Res. 2016;76(5):1181–92.

    Article  CAS  PubMed  Google Scholar 

  74. Li S, Zhao H, Fan Y, Zhao G, Wang R, Wen F, Wang J, Wang X, Wang Y, Gao Y. Design, synthesis, and in vitro antitumor activity of a transferrin receptor-targeted peptide-doxorubicin conjugate. Chem Biol Drug Des. 2020;95(1):58–65.

    Article  CAS  PubMed  Google Scholar 

  75. Gowland C, Berry P, Errington J, Jeffrey P, Bennett G, Godfrey L, Pittman M, Niewiarowski A, Symeonides SN, Veal GJ. Development of a LC-MS/MS method for the quantification of toxic payload DM1 cleaved from BT1718 in a phase I study. Bioanalysis. 2021;13(2):101–13.

    Article  CAS  PubMed  Google Scholar 

  76. Chen L, Zeng D, Xu N, Li C, Zhang W, Zhu X, Gao Y, Chen PR, Lin J. Blood-Brain Barrier- and Blood-Brain tumor Barrier-Penetrating Peptide-Derived targeted therapeutics for glioma and malignant tumor brain metastases. ACS Appl Mater Interfaces. 2019;11(45):41889–97.

    Article  CAS  PubMed  Google Scholar 

  77. Kumthekar P, Tang SC, Brenner AJ, Kesari S, Piccioni DE, Anders C, Carrillo J, Chalasani P, Kabos P, Puhalla S, Tkaczuk K, Garcia AA, Ahluwalia MS, Wefel JS, Lakhani N, Ibrahim N. ANG1005, a brain-Penetrating Peptide-Drug conjugate, shows activity in patients with breast Cancer with leptomeningeal carcinomatosis and recurrent brain metastases. Clin Cancer Res. 2020;26(12):2789–99.

    Article  CAS  PubMed  Google Scholar 

  78. Che C, Yang G, Thiot C, Lacoste MC, Currie JC, Demeule M, Regina A, Beliveau R, Castaigne JP. New Angiopep-modified doxorubicin (ANG1007) and Etoposide (ANG1009) chemotherapeutics with increased brain penetration. J Med Chem. 2010;53(7):2814–24.

    Article  CAS  PubMed  Google Scholar 

  79. Wang S, Noberini R, Stebbins JL, Das S, Zhang Z, Wu B, Mitra S, Billet S, Fernandez A, Bhowmick NA, Kitada S, Pasquale EB, Fisher PB, Pellecchia M. Targeted delivery of Paclitaxel to EphA2-expressing cancer cells. Clin Cancer Res. 2013;19(1):128–37.

    Article  CAS  PubMed  Google Scholar 

  80. Mudd GE, Brown A, Chen L, van Rietschoten K, Watcham S, Teufel DP, Pavan S, Lani R, Huxley P, Bennett GS. Identification and optimization of EphA2-Selective bicycles for the delivery of cytotoxic payloads. J Med Chem. 2020;63(8):4107–16.

    Article  CAS  PubMed  Google Scholar 

  81. Barile E, Wang S, Das SK, Noberini R, Dahl R, Stebbins JL, Pasquale EB, Fisher PB, Pellecchia M. Design, synthesis and bioevaluation of an EphA2 receptor-based targeted delivery system. ChemMedChem. 2014;9(7):1403–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Fan M, Yang D, Liang X, Ao J, Li Z, Wang H, Shi B. Design and biological activity of epidermal growth factor receptor-targeted peptide doxorubicin conjugate. Biomed Pharmacother. 2015;70:268–73.

    Article  CAS  PubMed  Google Scholar 

  83. Ai S, Duan J, Liu X, Bock S, Tian Y, Huang Z. Biological evaluation of a novel doxorubicin-peptide conjugate for targeted delivery to EGF receptor-overexpressing tumor cells. Mol Pharm. 2011;8(2):375–86.

    Article  CAS  PubMed  Google Scholar 

  84. Toyama K, Nomura W, Kobayakawa T, Tamamura H. Delivery of a proapoptotic peptide to EGFR-Positive Cancer cells by a Cyclic peptide mimicking the dimerization arm structure of EGFR. Bioconjug Chem. 2018;29(6):2050–7.

    Article  CAS  PubMed  Google Scholar 

  85. Li L, Li JC, Yang H, Zhang X, Liu LL, Li Y, Zeng TT, Zhu YH, Li XD, Li Y, Xie D, Fu L, Guan XY. Expansion of cancer stem cell pool initiates lung cancer recurrence before angiogenesis. Proc Natl Acad Sci U S A. 2018;115(38):E8948–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Sun S, Meng Y, Li M, Tang X, Hu W, Wu W, Li G, Pang Q, Wang W, Liu B. CD133(+) endothelial-like stem cells restore neovascularization and promote longevity in progeroid and naturally aged mice. Nat Aging. 2023;3(11):1401–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Sun J, Zhang C, Liu G, Liu H, Zhou C, Lu Y, Zhou C, Yuan L, Li X. A novel mouse CD133 binding-peptide screened by phage display inhibits cancer cell motility in vitro. Clin Exp Metastasis. 2012;29(3):185–96.

    Article  PubMed  Google Scholar 

  88. Li HM, Dong ZP, Wang QY, Liu LX, Li BX, Ma XN, Lin MS, Lu T, Wang Y. De Novo computational design for development of a peptide ligand oriented to VEGFR-3 with high affinity and long circulation. Mol Pharm. 2017;14(7):2236–44.

    Article  CAS  PubMed  Google Scholar 

  89. Wang QY, Li HM, Dong ZP, Li BX, Huo M, Lu T, Wang Y. Peptide-mediated cationic micelles drug-delivery system applied on a VEGFR3-overexpressed tumor. J Mater Chem B. 2019;7(7):1076–86.

    Article  CAS  PubMed  Google Scholar 

  90. Ying M, Shen Q, Zhan C, Wei X, Gao J, Xie C, Yao B, Lu W. A stabilized peptide ligand for multifunctional glioma targeted drug delivery. J Control Release. 2016;243:86–98.

    Article  CAS  PubMed  Google Scholar 

  91. Wei SC, Duffy CR, Allison JP. Fundamental mechanisms of immune checkpoint Blockade therapy. Cancer Discov. 2018;8(9):1069–86.

    Article  PubMed  Google Scholar 

  92. Sun Y, Lyu B, Yang C, He B, Zhang H, Wang X, Zhang Q, Dai W. An enzyme-responsive and transformable PD-L1 blocking peptide-photosensitizer conjugate enables efficient photothermal immunotherapy for breast cancer. Bioact Mater. 2023;22:47–59.

    CAS  PubMed  Google Scholar 

  93. Zeng Z, Yang Z, Li C, Liu S, Wei W, Zhou Y, Wang S, Sui M, Li M, Lin S, Cheng Y, Hou P. Advancing Cancer immunotherapy through engineering new PD-L1 degraders: A comprehensive study from small molecules to PD-L1-Specific Peptide-Drug conjugates. J Med Chem. 2024;67(21):19216–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Wang M, Shu XS, Li M, Zhang Y, Yao Y, Huang X, Li J, Wei P, He Z, Lu J, Ying Y. A novel strategy conjugating PD-L1 polypeptide with doxorubicin alleviates chemotherapeutic resistance and enhances immune response in Colon cancer. Front Oncol. 2021;11:737323.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Feng M, Jiang W, Kim BYS, Zhang CC, Fu YX, Weissman IL. Phagocytosis checkpoints as new targets for cancer immunotherapy. Nat Rev Cancer. 2019;19(10):568–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Ehrlich GK, Michel H, Truitt T, Riboulet W, Pop-Damkov P, Goelzer P, Hainzl D, Qureshi F, Lueckel B, Danho W, Conde-Knape K, Konkar A. Preparation and characterization of albumin conjugates of a truncated peptide YY analogue for half-life extension. Bioconjug Chem. 2013;24(12):2015–24.

    Article  CAS  PubMed  Google Scholar 

  97. Kurrikoff K, Gestin M, Langel U. Recent in vivo advances in cell-penetrating peptide-assisted drug delivery. Expert Opin Drug Deliv. 2016;13(3):373–87.

    Article  CAS  PubMed  Google Scholar 

  98. Nguyen DD, Luo LJ, Yang CJ, Lai JY. Highly Retina-Permeating and Long-Acting Resveratrol/Metformin nanotherapeutics for enhanced treatment of macular degeneration. ACS Nano. 2023;17(1):168–83.

    Article  CAS  PubMed  Google Scholar 

  99. Shi M, Jiang Z, Xiao Y, Song Y, Tang R, Zhang L, Huang J, Tian Y, Zhou S. Stapling of short cell-penetrating peptides for enhanced tumor cell-and-tissue dual-penetration. Chem Commun (Camb). 2022;58(14):2299–302.

    Article  CAS  PubMed  Google Scholar 

  100. Tian Y, Zeng X, Li J, Jiang Y, Zhao H, Wang D, Huang X, Li Z. Achieving enhanced cell penetration of short conformationally constrained peptides through amphiphilicity tuning. Chem Sci. 2017;8(11):7576–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Chen Z, Zhang P, Cheetham AG, Moon JH, Moxley JW Jr., Lin YA, Cui H. Controlled release of free doxorubicin from peptide-drug conjugates by drug loading. J Control Release. 2014;191:123–30.

    Article  CAS  PubMed  Google Scholar 

  102. Hu G, Guo M, Xu J, Wu F, Fan J, Huang Q, Yang G, Lv Z, Wang X, Jin Y. Nanoparticles Targeting Macrophages as Potential Clinical Therapeutic Agents Against Cancer and Inflammation, Front Immunol 10 (2019) 1998.

  103. Gottesman MM, Fojo T, Bates SE. Multidrug resistance in cancer: role of ATP-dependent transporters. Nat Rev Cancer. 2002;2(1):48–58.

    Article  CAS  PubMed  Google Scholar 

  104. Pauwels EK, Erba P, Mariani G, Gomes CM. Multidrug resistance in cancer: its mechanism and its modulation. Drug News Perspect. 2007;20(6):371–7.

    Article  CAS  PubMed  Google Scholar 

  105. Stanzl EG, Trantow BM, Vargas JR, Wender PA. Fifteen years of cell-penetrating, guanidinium-rich molecular transporters: basic science, research tools, and clinical applications. Acc Chem Res. 2013;46(12):2944–54.

    Article  CAS  PubMed  Google Scholar 

  106. Cheng H, Zhu JY, Xu XD, Qiu WX, Lei Q, Han K, Cheng YJ, Zhang XZ. Activable Cell-Penetrating peptide conjugated prodrug for tumor targeted drug delivery. ACS Appl Mater Interfaces. 2015;7(29):16061–9.

    Article  CAS  PubMed  Google Scholar 

  107. Wang D, Li W, Zhao R, Chen L, Liu N, Tian Y, Zhao H, Xie M, Lu F, Fang Q, Liang W, Yin F, Li Z. Stabilized peptide HDAC inhibitors derived from HDAC1 substrate H3K56 for the treatment of Cancer Stem-Like cells in vivo. Cancer Res. 2019;79(8):1769–83.

    Article  CAS  PubMed  Google Scholar 

  108. Du L, Song C, Du J, Zeng F, Zhang Y, Yin F, Li Z, Wang D. Development of sulfonium tethered peptides conjugated with HDAC inhibitor to improve selective toxicity for cancer cells. Bioorg Med Chem. 2023;83:117213.

    Article  CAS  PubMed  Google Scholar 

  109. Otter R, Besenius P. Supramolecular assembly of functional peptide-polymer conjugates. Org Biomol Chem. 2019;17(28):6719–34.

    Article  CAS  PubMed  Google Scholar 

  110. Aida T, Meijer EW, Stupp SI. Functional supramolecular polymers. Science. 2012;335(6070):813–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Matson JB, Stupp SI. Drug release from hydrazone-containing peptide amphiphiles. Chem Commun (Camb). 2011;47(28):7962–4.

    Article  CAS  PubMed  Google Scholar 

  112. Gao Y, Kuang Y, Guo ZF, Guo Z, Krauss IJ, Xu B. Enzyme-instructed molecular self-assembly confers nanofibers and a supramolecular hydrogel of taxol derivative. J Am Chem Soc. 2009;131(38):13576–7.

    Article  CAS  PubMed  Google Scholar 

  113. Li J, Gao Y, Kuang Y, Shi J, Du X, Zhou J, Wang H, Yang Z, Xu B. Dephosphorylation of D-peptide derivatives to form biofunctional, supramolecular nanofibers/hydrogels and their potential applications for intracellular imaging and intratumoral chemotherapy. J Am Chem Soc. 2013;135(26):9907–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Lock LL, Tang Z, Keith D, Reyes C, Cui H. Enzyme-Specific doxorubicin drug beacon as drug-Resistant theranostic molecular probes. ACS Macro Lett. 2015;4(5):552–5.

    Article  CAS  PubMed  Google Scholar 

  115. Zhang P, Cheetham AG, Lin YA, Cui H. Self-assembled Tat nanofibers as effective drug carrier and transporter. ACS Nano. 2013;7(7):5965–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Wang D, Tian Y, Zhang Y, Sun X, Wu Y, Liu R, Zeng F, Du J, Hu K. An assembly-inducing PDC enabling the efficient nuclear delivery of nucleic acid for cancer stem-like cell suppression. Nanoscale. 2022;14(41):15384–92.

    Article  CAS  PubMed  Google Scholar 

  117. Zhang Y, He P, Zhang P, Yi X, Xiao C, Chen X. Polypeptides-Drug conjugates for anticancer therapy. Adv Healthc Mater. 2021;10(11):e2001974.

    Article  PubMed  Google Scholar 

  118. Bae Y, Fukushima S, Harada A, Kataoka K. Design of environment-sensitive supramolecular assemblies for intracellular drug delivery: polymeric micelles that are responsive to intracellular pH change. Angew Chem Int Ed Engl. 2003;42(38):4640–3.

    Article  CAS  PubMed  Google Scholar 

  119. Cai C, Li Y, Lin J, Wang L, Lin S, Wang XS, Jiang T. Simulation-assisted self-assembly of multicomponent polymers into hierarchical assemblies with varied morphologies. Angew Chem Int Ed Engl. 2013;52(30):7732–6.

    Article  CAS  PubMed  Google Scholar 

  120. Cai C, Lin J, Lu Y, Zhang Q, Wang L. Polypeptide self-assemblies: nanostructures and bioapplications. Chem Soc Rev. 2016;45(21):5985–6012.

    Article  CAS  PubMed  Google Scholar 

  121. Bhargava P, Tu Y, Zheng JX, Xiong H, Quirk RP, Cheng SZ. Temperature-induced reversible morphological changes of polystyrene-block-poly(ethylene oxide) micelles in solution. J Am Chem Soc. 2007;129(5):1113–21.

    Article  CAS  PubMed  Google Scholar 

  122. Sis MJ, Ye Z, La Costa K, Webber MJ. Energy landscapes of supramolecular Peptide-Drug conjugates directed by linker selection and drug topology. ACS Nano. 2022;16(6):9546–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. You Y, Xu Z, Chen Y. Doxorubicin conjugated with a trastuzumab epitope and an MMP-2 sensitive peptide linker for the treatment of HER2-positive breast cancer. Drug Deliv. 2018;25(1):448–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Bajjuri KM, Liu Y, Liu C, Sinha SC. The legumain protease-activated auristatin prodrugs suppress tumor growth and metastasis without toxicity. ChemMedChem. 2011;6(1):54–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Huang TL, Szekacs A, Uematsu T, Kuwano E, Parkinson A, Hammock BD. Hydrolysis of carbonates, thiocarbonates, carbamates, and carboxylic esters of alpha-naphthol, beta-naphthol, and p-nitrophenol by human, rat, and mouse liver carboxylesterases. Pharm Res. 1993;10(5):639–48.

    Article  CAS  PubMed  Google Scholar 

  126. Laizure SC, Herring V, Hu Z, Witbrodt K, Parker RB. The role of human carboxylesterases in drug metabolism: have we overlooked their importance? Pharmacotherapy. 2013;33(2):210–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Vacondio F, Silva C, Mor M, Testa B. Qualitative structure-metabolism relationships in the hydrolysis of carbamates. Drug Metab Rev. 2010;42(4):551–89.

    Article  CAS  PubMed  Google Scholar 

  128. Karampelas T, Argyros O, Sayyad N, Spyridaki K, Pappas C, Morgan K, Kolios G, Millar RP, Liapakis G, Tzakos AG, Fokas D, Tamvakopoulos C. GnRH-Gemcitabine conjugates for the treatment of androgen-independent prostate cancer: Pharmacokinetic enhancements combined with targeted drug delivery. Bioconjug Chem 25(4) (2014) 813– 23.

  129. Sayyad N, Vrettos EI, Karampelas T, Chatzigiannis CM, Spyridaki K, Liapakis G, Tamvakopoulos C, Tzakos AG. Development of bioactive gemcitabine-D-Lys(6)-GnRH prodrugs with linker-controllable drug release rate and enhanced biopharmaceutical profile. Eur J Med Chem. 2019;166:256–66.

    Article  CAS  PubMed  Google Scholar 

  130. Cho YS, Kim GC, Lee HM, Kim B, Kim HR, Chung SW, Chang HW, Ko YG, Lee YS, Kim SW, Byun Y, Kim SY. Albumin metabolism targeted peptide-drug conjugate strategy for targeting pan-KRAS mutant cancer. J Control Release. 2022;344:26–38.

    Article  CAS  PubMed  Google Scholar 

  131. Cho YS, Cho H, Kim HR, Park SJ, Yeo JH, Ko YG, Lee J, Kim SY, Kim K, Byun Y. Macropinocytosis-targeted peptide-docetaxel conjugate for bystander pancreatic cancer treatment. J Control Release. 2024;376:829–41.

    Article  CAS  PubMed  Google Scholar 

  132. Bargh JD, Isidro-Llobet A, Parker JS, Spring DR. Cleavable linkers in antibody-drug conjugates. Chem Soc Rev. 2019;48(16):4361–74.

    Article  CAS  PubMed  Google Scholar 

  133. Ziaei E, Saghaeidehkordi A, Dill C, Maslennikov I, Chen S, Kaur K. Targeting triple negative breast Cancer cells with novel cytotoxic Peptide-Doxorubicin conjugates. Bioconjug Chem. 2019;30(12):3098–106.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Mollaev M, Gorokhovets N, Nikolskaya E, Faustova M, Zabolotsky A, Zhunina O, Sokol M, Zamulaeva I, Severin E, Yabbarov N. Type of pH sensitive linker reveals different time-dependent intracellular localization, in vitro and in vivo efficiency in alpha-fetoprotein receptor targeted doxorubicin conjugate. Int J Pharm. 2019;559:138–46.

    Article  CAS  PubMed  Google Scholar 

  135. Tomlinson R, Heller J, Brocchini S, Duncan R. Polyacetal-doxorubicin conjugates designed for pH-dependent degradation. Bioconjug Chem. 2003;14(6):1096–106.

    Article  CAS  PubMed  Google Scholar 

  136. Sheng Y, You Y, Chen Y. Dual-targeting hybrid peptide-conjugated doxorubicin for drug resistance reversal in breast cancer. Int J Pharm. 2016;512(1):1–13.

    Article  CAS  PubMed  Google Scholar 

  137. Rizvi SFA, Abbas N, Zhang H, Fang Q. Identification of a pH-Responsive Peptide-Paclitaxel conjugate as a novel drug with improved therapeutic potential. J Med Chem. 2023;66(12):8324–37.

    Article  CAS  PubMed  Google Scholar 

  138. Li S, Zhao H, Mao X, Fan Y, Liang X, Wang R, Xiao L, Wang J, Liu Q, Zhao G. Transferrin receptor targeted cellular delivery of doxorubicin via a Reduction-Responsive Peptide-Drug conjugate. Pharm Res. 2019;36(12):168.

    Article  CAS  PubMed  Google Scholar 

  139. Fontaine SD, Reid R, Robinson L, Ashley GW, Santi DV. Long-term stabilization of maleimide-thiol conjugates. Bioconjug Chem. 2015;26(1):145–52.

    Article  CAS  PubMed  Google Scholar 

  140. Dokus LE, Lajko E, Randelovic I, Mezo D, Schlosser G, Kohidai L, Tovari J, Mezo G. Phage Display-Based Homing Peptide-Daunomycin Conjugates for Selective Drug Targeting to PANC-1 Pancreatic Cancer, Pharmaceutics 12(6) (2020).

  141. Zheng BY, Yang XQ, Zhao Y, Zheng QF, Ke MR, Lin T, Chen RX, Ho KKK, Kumar N, Huang JD. Synthesis and photodynamic activities of integrin-targeting silicon(IV) phthalocyanine-cRGD conjugates. Eur J Med Chem. 2018;155:24–33.

    Article  CAS  PubMed  Google Scholar 

  142. Schuster S, Biri-Kovacs B, Szeder B, Buday L, Gardi J, Szabo Z, Halmos G, Mezo G. Enhanced in vitro antitumor activity of GnRH-III-Daunorubicin bioconjugates influenced by sequence modification. Pharmaceutics 10(4) (2018).

  143. Randelovic I, Schuster S, Kapuvari B, Fossati G, Steinkuhler C, Mezo G, Tovari J. Improved in vivo Anti-Tumor and Anti-Metastatic effect of GnRH-III-Daunorubicin analogs on colorectal and breast carcinoma bearing mice. Int J Mol Sci 20(19) (2019).

  144. Duncan R. Polymer conjugates as anticancer nanomedicines. Nat Rev Cancer. 2006;6(9):688–701.

    Article  CAS  PubMed  Google Scholar 

  145. di Santo G, Santo G, Sviridenko A, Virgolini I. Peptide receptor radionuclide therapy combinations for neuroendocrine tumours in ongoing clinical trials: status 2023. Theranostics. 2024;14(3):940–53.

    Article  PubMed  PubMed Central  Google Scholar 

  146. Shao J, Lin X, Wang H, Zhao C, Yao SQ, Ge J, Zeng S, Qian L. Targeted degradation of Cell-Surface proteins via Chaperone-Mediated autophagy by using Peptide-Conjugated antibodies. Angew Chem Int Ed Engl. 2024;63(18):e202319232.

    Article  CAS  PubMed  Google Scholar 

  147. Rahman M, Sahoo A, Almalki WH, Almujri SS, Altamimi ASA, Alhamyani A, Akhter S. Peptide spiders are emerging as novel therapeutic interventions for nucleic acid delivery. Drug Discov Today. 2024;29(7):104021.

    Article  CAS  PubMed  Google Scholar 

  148. Shen W, Liu W, Yang H, Zhang P, Xiao C, Chen X. A glutathione-responsive sulfur dioxide polymer prodrug as a nanocarrier for combating drug-resistance in cancer chemotherapy. Biomaterials. 2018;178:706–19.

    Article  CAS  PubMed  Google Scholar 

  149. Zhang Y, Shen W, Zhang P, Chen L, Xiao C. GSH-triggered release of sulfur dioxide gas to regulate redox balance for enhanced photodynamic therapy. Chem Commun (Camb). 2020;56(42):5645–8.

    Article  CAS  PubMed  Google Scholar 

  150. Qian C, Wang J, Qian Y, Hu R, Zou J, Zhu C, Zhu Y, Qi S, Jia X, Wu L, Li W, Chen Z. Tumor-Cell-Surface adherable Peptide-Drug conjugate prodrug nanoparticles inhibit tumor metastasis and augment treatment efficacy. Nano Lett. 2020;20(6):4153–61.

    Article  CAS  PubMed  Google Scholar 

  151. Zoghebi K, Aliabadi HM, Tiwari RK, Parang K. [(WR)(8)WKbetaA]-Doxorubicin conjugate: A delivery system to overcome Multi-Drug resistance against doxorubicin. Cells 11(2) (2022).

  152. Hua D, Tang L, Wang W, Tang S, Yu L, Zhou X, Wang Q, Sun C, Shi C, Luo W, Jiang Z, Li H, Yu S. Improved antiglioblastoma activity and BBB permeability by conjugation of Paclitaxel to a Cell-Penetrative MMP-2-Cleavable peptide. Adv Sci (Weinh). 2021;8(3):2001960.

    Article  CAS  PubMed  Google Scholar 

  153. Guo XM, Yadav MB, Khan M, Hao CW, Lin CY, Huang T, Wu J, Fan BM, Bian ZX. Bradykinin-Potentiating Peptide-Paclitaxel conjugate directed at ectopically expressed Angiotensin-Converting enzyme in Triple-Negative breast Cancer. J Med Chem. 2021;64(23):17051–62.

    Article  CAS  PubMed  Google Scholar 

  154. Wang L, Chen H, Wang F, Zhang X. The development of peptide-drug conjugates (PDCs) strategies for Paclitaxel. Expert Opin Drug Deliv. 2022;19(2):147–61.

    Article  CAS  PubMed  Google Scholar 

  155. Zheng S, Cai Y, Hong Y, Gong Y, Gao L, Li Q, Li L, Sun X. Legumain/pH dual-responsive lytic peptide-paclitaxel conjugate for synergistic cancer therapy. Drug Deliv. 2022;29(1):1764–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Hou L, Hou Y, Liang Y, Chen B, Zhang X, Wang Y, Zhou K, Zhong T, Long B, Pang W, Wang L, Han X, Li L, Xu C, Gross I, Gaiddon C, Fu W, Yao H, Meng X. Anti-tumor effects of P-LPK-CPT, a peptide-camptothecin conjugate, in colorectal cancer. Commun Biol. 2022;5(1):1248.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Jimenez-Macias JL, Lee YC, Miller E, Finkelberg T, Zdioruk M, Berger G, Farquhar CE, Nowicki MO, Cho CF, Fedeles BI, Loas A, Pentelute BL, Lawler SE. A Pt(IV)-conjugated brain penetrant macrocyclic peptide shows pre-clinical efficacy in glioblastoma. J Control Release. 2022;352:623–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Kim MS, Ma S, Chelariu-Raicu A, Leuschner C, Alila HW, Lee S, Coleman RL, Sood AK. Enhanced immunotherapy with LHRH-R targeted lytic peptide in ovarian Cancer. Mol Cancer Ther. 2020;19(11):2396–406.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Alves ID, Carre M, Montero MP, Castano S, Lecomte S, Marquant R, Lecorche P, Burlina F, Schatz C, Sagan S, Chassaing G, Braguer D, Lavielle S. A proapoptotic peptide conjugated to penetratin selectively inhibits tumor cell growth. Biochim Biophys Acta. 2014;1838(8):2087–98.

    Article  CAS  PubMed  Google Scholar 

  160. Huang Y, Li X, Sha H, Zhang L, Bian X, Han X, Liu B. Tumor-penetrating peptide fused to a pro-apoptotic peptide facilitates effective gastric cancer therapy. Oncol Rep. 2017;37(4):2063–70.

    Article  CAS  PubMed  Google Scholar 

  161. Lee JW, Choi J, Choi Y, Kim K, Yang Y, Kim SH, Yoon HY, Kwon IC. Molecularly engineered SiRNA conjugates for tumor-targeted RNAi therapy. J Control Release. 2022;351:713–26.

    Article  CAS  PubMed  Google Scholar 

  162. Smidt JM, Lykke L, Stidsen CE, Pristovsek N, Gothelf KV. Synthesis of peptide-siRNA conjugates via internal sulfonylphosphoramidate modifications and evaluation of their in vitro activity. Nucleic Acids Res. 2024;52(1):49–58.

    Article  CAS  PubMed  Google Scholar 

  163. Leeson PD, Springthorpe B. The influence of drug-like concepts on decision-making in medicinal chemistry. Nat Rev Drug Discov. 2007;6(11):881–90.

    Article  CAS  PubMed  Google Scholar 

  164. Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev. 2001;46(1–3):3–26.

    Article  CAS  PubMed  Google Scholar 

  165. Yamamoto A. [Improvement of intestinal and transmucosal absorption of peptide and protein drugs]. Yakugaku Zasshi. 2024;144(7):697–714.

    Article  CAS  PubMed  Google Scholar 

  166. Kurtzhals P, Ostergaard S, Nishimura E, Kjeldsen T. Derivatization with fatty acids in peptide and protein drug discovery. Nat Rev Drug Discov. 2023;22(1):59–80.

    Article  CAS  PubMed  Google Scholar 

  167. Krecmerova M. Amino acid ester prodrugs of nucleoside and nucleotide antivirals. Mini Rev Med Chem. 2017;17(10):818–33.

    Article  CAS  PubMed  Google Scholar 

  168. Wiemer AJ, Wiemer DF. Prodrugs of phosphonates and phosphates: crossing the membrane barrier. Top Curr Chem. 2015;360:115–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Park SE, Sajid MI, Parang K, Tiwari RK. Cyclic Cell-Penetrating peptides as efficient intracellular drug delivery tools. Mol Pharm. 2019;16(9):3727–43.

    Article  CAS  PubMed  Google Scholar 

  170. Clark RJ, Jensen J, Nevin ST, Callaghan BP, Adams DJ, Craik DJ. The engineering of an orally active conotoxin for the treatment of neuropathic pain. Angew Chem Int Ed Engl. 2010;49(37):6545–8.

    Article  CAS  PubMed  Google Scholar 

  171. Chen G, Kang W, Li W, Chen S, Gao Y. Oral delivery of protein and peptide drugs: from non-specific formulation approaches to intestinal cell targeting strategies. Theranostics. 2022;12(3):1419–39.

    Article  PubMed  PubMed Central  Google Scholar 

  172. Kamei N, Morishita M, Ehara J, Takayama K. Permeation characteristics of oligoarginine through intestinal epithelium and its usefulness for intestinal peptide drug delivery. J Control Release. 2008;131(2):94–9.

    Article  CAS  PubMed  Google Scholar 

  173. Aguero L, Zaldivar-Silva D, Pena L, Dias ML. Alginate microparticles as oral colon drug delivery device: A review. Carbohydr Polym. 2017;168:32–43.

    Article  CAS  PubMed  Google Scholar 

  174. Carrillo-Conde BR, Brewer E, Lowman A, Peppas NA. Complexation hydrogels as oral delivery vehicles of therapeutic antibodies: an in vitro and ex vivo evaluation of antibody stability and bioactivity. Ind Eng Chem Res. 2015;54(42):10197–205.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Nielsen DS, Shepherd NE, Xu W, Lucke AJ, Stoermer MJ, Fairlie DP. Orally Absorbed Cycl Peptides Chem Rev. 2017;117(12):8094–128.

    CAS  Google Scholar 

  176. Fu C, Yu L, Miao Y, Liu X, Yu Z, Wei M. Peptide-drug conjugates (PDCs): a novel trend of research and development on targeted therapy, hype or hope? Acta Pharm Sin B. 2023;13(2):498–516.

    Article  CAS  PubMed  Google Scholar 

  177. Mayolo-Deloisa K, Gonzalez-Gonzalez M, Simental-Martinez J, Rito-Palomares M. Aldehyde pegylation of laccase from trametes versicolor in route to increase its stability: effect on enzymatic activity. J Mol Recognit. 2015;28(3):173–9.

    Article  CAS  PubMed  Google Scholar 

  178. Khan AA, Morrison A, Hanley DA, Felsenberg D, McCauley LK, O’Ryan F, Reid IR, Ruggiero SL, Taguchi A, Tetradis S, Watts NB, Brandi ML, Peters E, Guise T, Eastell R, Cheung AM, Morin SN, Masri B, Cooper C, Morgan SL, Obermayer-Pietsch B, Langdahl BL, Al R, Dabagh KS, Davison DL, Kendler GK, Sandor RG, Josse M, Bhandari M, El Rabbany DD, Pierroz R, Sulimani DP, Saunders JP, Brown J, Compston J. International task force on osteonecrosis of the, diagnosis and management of osteonecrosis of the jaw: a systematic review and international consensus. J Bone Min Res. 2015;30(1):3–23.

    Article  Google Scholar 

  179. Lau J, Bloch P, Schaffer L, Pettersson I, Spetzler J, Kofoed J, Madsen K, Knudsen LB, McGuire J, Steensgaard DB, Strauss HM, Gram DX, Knudsen SM, Nielsen FS, Thygesen P, Reedtz-Runge S, Kruse T. Discovery of the Once-Weekly Glucagon-Like Peptide-1 (GLP-1) analogue semaglutide. J Med Chem. 2015;58(18):7370–80.

    Article  CAS  PubMed  Google Scholar 

  180. Garai I, Barna S, Nagy G, Forgacs A. Limitations and pitfalls of 99mTc-EDDA/HYNIC-TOC (Tektrotyd) scintigraphy. Nucl Med Rev Cent East Eur. 2016;19(2):93–8.

    Article  PubMed  Google Scholar 

  181. Eastman KAS, Kincannon WM, Bandarian V. Leveraging substrate promiscuity of a radical S-Adenosyl-L-methionine RiPP maturase toward intramolecular peptide Cross-Linking applications. ACS Cent Sci. 2022;8(8):1209–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Strosberg JR, Caplin ME, Kunz PL, Ruszniewski PB, Bodei L, Hendifar A, Mittra E, Wolin EM, Yao JC, Pavel ME, Grande E, Van Cutsem E, Seregni E, Duarte H, Gericke G, Bartalotta A, Mariani MF, Demange A, Mutevelic S, Krenning EP, investigators N.-. (177)Lu-Dotatate plus long-acting octreotide versus high–dose long-acting octreotide in patients with midgut neuroendocrine tumours (NETTER-1): final overall survival and long-term safety results from an open-label, randomised, controlled, phase 3 trial. Lancet Oncol. 2021;22(12):1752–63.

    Article  CAS  PubMed  Google Scholar 

  183. Menda Y, O’Dorisio TM, Howe JR, Schultz M, Dillon JS, Dick D, Watkins GL, Ginader T, Bushnell DL, Sunderland JJ, Zamba GKD, Graham M. O’Dorisio, localization of unknown primary site with (68)Ga-DOTATOC PET/CT in patients with metastatic neuroendocrine tumor. J Nucl Med. 2017;58(7):1054–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Ambrosini V, Fanti S. Radioguided surgery with 68Ga-DOTATATE for patients with neuroendocrine tumors. Hepatobiliary Surg Nutr. 2020;9(1):67–9.

    Article  PubMed  PubMed Central  Google Scholar 

  185. Yu SS, Athreya K, Liu SV, Schally AV, Tsao-Wei D, Groshen S, Quinn DI, Dorff TB, Xiong S, Engel J, Pinski J. A phase II trial of AEZS-108 in Castration- and Taxane-Resistant prostate Cancer. Clin Genitourin Cancer. 2017;15(6):742–9.

    Article  PubMed  Google Scholar 

  186. Emons G, Gorchev G, Harter P, Wimberger P, Stahle A, Hanker L, Hilpert F, Beckmann MW, Dall P, Grundker C, Sindermann H, Sehouli J. Efficacy and safety of AEZS-108 (LHRH agonist linked to doxorubicin) in women with advanced or recurrent endometrial cancer expressing LHRH receptors: a multicenter phase 2 trial (AGO-GYN5). Int J Gynecol Cancer. 2014;24(2):260–5.

    Article  PubMed  PubMed Central  Google Scholar 

  187. Drappatz J, Brenner A, Wong ET, Eichler A, Schiff D, Groves MD, Mikkelsen T, Rosenfeld S, Sarantopoulos J, Meyers CA, Fielding RM, Elian K, Wang X, Lawrence B, Shing M, Kelsey S, Castaigne JP, Wen PY. Phase I study of GRN1005 in recurrent malignant glioma. Clin Cancer Res. 2013;19(6):1567–76.

    Article  CAS  PubMed  Google Scholar 

  188. Jo U, Murai Y, Agama KK, Sun Y, Saha LK, Yang X, Arakawa Y, Gayle S, Jones K, Paralkar V, Sundaram RK, Van Doorn J, Vasquez JC, Bindra RS, Choi WS, Pommier Y. TOP1-DNA trapping by Exatecan and combination therapy with ATR inhibitor. Mol Cancer Ther. 2022;21(7):1090–102.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Wyatt LC, Moshnikova A, Crawford T, Engelman DM, Andreev OA, Reshetnyak YK. Peptides of pHLIP family for targeted intracellular and extracellular delivery of cargo molecules to tumors. Proc Natl Acad Sci U S A. 2018;115(12):E2811–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Gayle S, Aiello R, Leelatian N, Beckta JM, Bechtold J, Bourassa P, Csengery J, Maguire RJ, Marshall D, Sundaram RK, Van Doorn J, Jones K, Moore H, Lopresti-Morrow L, Paradis T, Tylaska L, Zhang Q, Visca H, Reshetnyak YK, Andreev OA, Engelman DM, Glazer PM, Bindra RS, Paralkar VM. Tumor-selective, antigen-independent delivery of a pH sensitive peptide-topoisomerase inhibitor conjugate suppresses tumor growth without systemic toxicity. NAR Cancer. 2021;3(2):zcab021.

    Article  PubMed  PubMed Central  Google Scholar 

  191. Demeule M, Charfi C, Currie JC, Larocque A, Zgheib A, Kozelko S, Beliveau R, Marsolais C, Annabi B. TH1902, a new docetaxel-peptide conjugate for the treatment of sortilin-positive triple-negative breast cancer. Cancer Sci. 2021;112(10):4317–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Bertrand N, Mougel R, Riley G, Bruand M, Gauchotte G, Agopiantz M. Neurotensin and its involvement in female Hormone-Sensitive cancers. Int J Mol Sci 25(21) (2024).

  193. Gao Y, Li Y, Song Z, Jin Z, Li X, Yuan C. Sortilin 1 promotes hepatocellular carcinoma cell proliferation and migration by regulating immune cell infiltration. J Oncol. 2022;2022:6509028.

    Article  PubMed  PubMed Central  Google Scholar 

  194. Demeule M, Charfi C, Currie JC, Zgheib A, Danalache BA, Beliveau R, Marsolais C, Annabi B. The TH1902 docetaxel Peptide-Drug conjugate inhibits xenografts growth of human SORT1-Positive ovarian and Triple-Negative breast Cancer Stem-like cells. Pharmaceutics 14(9) (2022).

  195. Chelariu-Raicu A, Nick A, Urban R, Gordinier M, Leuschner C, Bavisotto L, Molin GZD, Whisnant JK, Coleman RL. A multicenter open-label randomized phase II trial of Paclitaxel plus EP-100, a novel LHRH receptor-targeted, membrane-disrupting peptide, versus Paclitaxel alone for refractory or recurrent ovarian cancer. Gynecol Oncol. 2021;160(2):418–26.

    Article  CAS  PubMed  Google Scholar 

  196. Curtis KK, Sarantopoulos J, Northfelt DW, Weiss GJ, Barnhart KM, Whisnant JK, Leuschner C, Alila H, Borad MJ, Ramanathan RK. Novel LHRH-receptor-targeted cytolytic peptide, EP-100: first-in-human phase I study in patients with advanced LHRH-receptor-expressing solid tumors. Cancer Chemother Pharmacol. 2014;73(5):931–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Bashir B, Wang JS, Falchook G, Fontana E, Arkenau HT, Carter L, Galot R, Basu B, Greystoke A, Subbiah V, Richardson DL, Orr H, Bennett G, Sharma R, Xu H, Paganoni P, Xu C, Campbell C, McKean M. Results from First-in-Human phase I Dose-Escalation study of a novel bicycle toxin conjugate targeting EphA2 (BT5528) in patients with advanced solid tumors. J Clin Oncol. 2024;42(29):3443–52.

    Article  CAS  PubMed  Google Scholar 

  198. Mudd GE, Scott H, Chen L, van Rietschoten K, Ivanova-Berndt G, Dzionek K, Brown A, Watcham S, White L, Park PU, Jeffrey P, Rigby M, Beswick P. Discovery of BT8009: A Nectin-4 targeting bicycle toxin conjugate for the treatment of Cancer. J Med Chem. 2022;65(21):14337–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Cook N, Banerji U, Evans J, Biondo A, Germetaki T, Randhawa M, Godfrey L, Leslie S, Jeffrey P, Rigby M, Bennett G, Blakemore S, Koehler M, Niewiarowski A, Pittman M, Symeonides SN. Pharmacokinetic (PK) assessment of BT1718: A phase I/II a study of BT1718, a first in class bicycle toxin conjugate (BTC), in patients (pts) with advanced solid tumours. Ann Oncol. 2019;30:174–174.

    Article  Google Scholar 

  200. Bashir B, Wang JS, Falchook G, Fontana E, Arkenau HT, Carter L, Galot R, Basu B, Greystoke A, Subbiah V, Richardson DL, Orr H, Bennett G, Sharma R, Xu HM, Paganoni P, Xu C, Campbell C, McKean M. Results from First-in-Human phase I Dose-Escalation study of a novel bicycle toxin conjugate targeting EphA2 (BT5528) in patients with advanced solid tumors. J Clin Oncol 42(29) (2024).

  201. Bendell JC, Wang JSZ, Bashir B, Richardson DL, Bennett G, Campbell C, Hennessy MG, Jeffrey P, Kirui J, Mahnke L, Shapiro G. BT5528-100 phase I/II study of the safety, pharmacokinetics, and preliminary clinical activity of BT5528 in patients with advanced malignancies associated with EphA2 expression. J Clin Oncol 38(15) (2020).

  202. Puri S, Mazza M, Roy G, England RM, Zhou L, Nourian S. Anand subramony, evolution of nanomedicine formulations for targeted delivery and controlled release. Adv Drug Deliv Rev. 2023;200:114962.

    Article  CAS  PubMed  Google Scholar 

  203. Rinke A, Wittenberg M, Schade-Brittinger C, Aminossadati B, Ronicke E, Gress TM, Muller HH, Arnold R, Group PS. Placebo-Controlled, Double-Blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors (PROMID): results of Long-Term survival. Neuroendocrinology. 2017;104(1):26–32.

    Article  CAS  PubMed  Google Scholar 

  204. Singer JW. Paclitaxel poliglumex (XYOTAX, CT-2103): a macromolecular taxane. J Control Release. 2005;109(1–3):120–6.

    Article  CAS  PubMed  Google Scholar 

  205. Wu C, Wang M, Sun J, Jia Y, Zhu X, Liu G, Zhu Y, Guan Y, Zhang Z, Pang X. Peptide-drug co-assembling: A potent armament against cancer. Theranostics. 2023;13(15):5322–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Morrison C. Constrained peptides’ time to Shine?? Nat Rev Drug Discov. 2018;17(8):531–3.

    Article  CAS  PubMed  Google Scholar 

  207. Reguera L, Rivera DG. Multicomponent reaction toolbox for peptide macrocyclization and stapling. Chem Rev. 2019;119(17):9836–60.

    Article  CAS  PubMed  Google Scholar 

  208. Fadzen CM, Wolfe JM, Cho CF, Chiocca EA, Lawler SE, Pentelute BL. Perfluoroarene-Based peptide macrocycles to enhance penetration across the Blood-Brain barrier. J Am Chem Soc. 2017;139(44):15628–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Wang Y, Li Y, Cao J, Meng Q, Li X, Zhang Y, Lam KS, Hong A, Liu R, Chen X. Development and characterization of a novel Peptide-Drug conjugate with DM1 for treatment of FGFR2-Positive tumors. Biomedicines 9(8) (2021).

  210. Huang L, Xie J, Bi Q, Li Z, Liu S, Shen Q, Li C. Highly selective targeting of hepatic stellate cells for liver fibrosis treatment using a d-Enantiomeric peptide ligand of Fn14 identified by Mirror-Image mRNA display. Mol Pharm. 2017;14(5):1742–53.

    Article  CAS  PubMed  Google Scholar 

  211. Wang W, Khojasteh SC, Su D. Biosynthetic strategies for macrocyclic peptides. Molecules 26(11) (2021).

  212. Yu Z, Xiao Z, Guan L, Bao P, Yu Y, Liang Y, Li M, Huang Z, Chen X, Chen R, Su Y, Ge J. Translocation of gasdermin D induced mitochondrial injury and mitophagy mediated quality control in lipopolysaccharide related cardiomyocyte injury. Clin Transl Med. 2022;12(8):e1002.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Yang CJ, Nguyen DD, Lai JY. Poly(l-Histidine)-Mediated On-Demand therapeutic delivery of roughened ceria nanocages for treatment of chemical eye injury. Adv Sci (Weinh). 2023;10(26):e2302174.

    Article  PubMed  Google Scholar 

  214. Yang CJ, Anand A, Huang CC, Lai JY. Unveiling the power of Gabapentin-Loaded nanoceria with multiple therapeutic capabilities for the treatment of dry eye disease. ACS Nano. 2023;17(24):25118–35.

    Article  CAS  PubMed  Google Scholar 

  215. Ghosh S, Su YH, Yang CJ, Lai JY. Design of highly adhesive Urchin-Like gold nanostructures for effective topical drug administration and symptomatic relief of corneal dryness. Small Struct 6(2) (2025).

  216. Ji X, Nielsen AL, Heinis C. Cyclic peptides for drug development. Angew Chem Int Ed Engl. 2024;63(3):e202308251.

    Article  CAS  PubMed  Google Scholar 

  217. Chen S, Rentero Rebollo I, Buth SA, Morales-Sanfrutos J, Touati J, Leiman PG, Heinis C. Bicyclic peptide ligands pulled out of cysteine-rich peptide libraries. J Am Chem Soc. 2013;135(17):6562–9.

    Article  CAS  PubMed  Google Scholar 

  218. Li X, Craven TW, Levine PM. Cyclic peptide screening methods for preclinical drug discovery. J Med Chem. 2022;65(18):11913–26.

    Article  CAS  PubMed  Google Scholar 

  219. Wendt M, Bellavita R, Gerber A, Efrem NL, van Ramshorst T, Pearce NM, Davey PRJ, Everard I, Vazquez-Chantada M, Chiarparin E, Grieco P, Hennig S, Grossmann TN. Bicyclic beta-Sheet mimetics that target the transcriptional coactivator beta-Catenin and inhibit Wnt signaling. Angew Chem Int Ed Engl. 2021;60(25):13937–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  220. Sathe RY, Bharatam PV. Drug-dendrimer complexes and conjugates: detailed furtherance through theory and experiments. Adv Colloid Interface Sci. 2022;303:102639.

    Article  CAS  PubMed  Google Scholar 

  221. Wang X, Zhang M, Li Y, Cong H, Yu B, Shen Y. Research status of dendrimer micelles in tumor therapy for drug delivery. Small. 2023;19(50):e2304006.

    Article  PubMed  Google Scholar 

  222. Sapra R, Verma RP, Maurya GP, Dhawan S, Babu J, Haridas V. Designer peptide and protein dendrimers: A Cross-Sectional analysis. Chem Rev. 2019;119(21):11391–441.

    Article  CAS  PubMed  Google Scholar 

  223. Sheikh A, Md S, Kesharwani P. RGD engineered dendrimer nanotherapeutic as an emerging targeted approach in cancer therapy. J Control Release. 2021;340:221–42.

    Article  CAS  PubMed  Google Scholar 

  224. Li N, Li N, Yi Q, Luo K, Guo C, Pan D, Gu Z. Amphiphilic peptide dendritic copolymer-doxorubicin nanoscale conjugate self-assembled to enzyme-responsive anti-cancer agent. Biomaterials. 2014;35(35):9529–45.

    Article  CAS  PubMed  Google Scholar 

  225. Chen J, Zou X. Self-assemble peptide biomaterials and their biomedical applications. Bioact Mater. 2019;4:120–31.

    PubMed  PubMed Central  Google Scholar 

  226. Li C, Lang J, Wang Y, Cheng Z, Zu M, Li F, Sun J, Deng Y, Ji T, Nie G, Zhao Y. Self-assembly of CXCR4 antagonist peptide-docetaxel conjugates for breast tumor multi-organ metastasis Inhibition. Acta Pharm Sin B. 2023;13(9):3849–61.

    Article  PubMed  PubMed Central  Google Scholar 

  227. Lamson NG, Berger A, Fein KC, Whitehead KA. Anionic nanoparticles enable the oral delivery of proteins by enhancing intestinal permeability. Nat Biomed Eng. 2020;4(1):84–96.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to express our gratitude to the National Natural Science Foundation of China.

Funding

This work was supported by National Natural Science Foundation of China (22007033, 82274026) and National Key Research and Development Program of China (2023YFF1205000).

Author information

Authors and Affiliations

Authors

Contributions

Dongyuan Wang: Writing original draft, conceptualization and revision. Chen Shi: Supervision, review and revision. Yu Zhang: Review and revision; Zigang Li: conceptualization, review and revision. Feng Yin: Review and revision.

Corresponding authors

Correspondence to Zigang Li, Yu Zhang or Chen Shi.

Ethics declarations

Ethics and consent to publish

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, D., Yin, F., Li, Z. et al. Current progress and remaining challenges of peptide–drug conjugates (PDCs): next generation of antibody-drug conjugates (ADCs)?. J Nanobiotechnol 23, 305 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12951-025-03277-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12951-025-03277-2

Keywords