Abstract
Pediatric brain tumors represent an exceptionally formidable and multifaceted challenge within the realm of oncology, standing as the preeminent cause of cancer-related mortality among children and adolescents. The landscape of these malignancies is remarkably heterogeneous, encompassing a diverse spectrum of histopathological types and molecular profiles, each with unique biological behaviors and clinical trajectories. Conventional therapeutic modalities, primarily comprising neurosurgical resection, highly focused radiation therapy, and systemic chemotherapy, frequently encounter considerable limitations. These limitations stem not only from the inherent biological aggressiveness and infiltrative nature of many pediatric brain tumors but also from their anatomical location within the exquisitely sensitive developing central nervous system (CNS). Consequently, while these treatments can be life-saving for some, they often yield suboptimal long-term survival rates for others, and are invariably associated with a substantial burden of debilitating acute and chronic neurocognitive, endocrine, and neurological sequelae that profoundly impact the quality of life for survivors. In light of these critical unmet needs, recent and ongoing advancements in the field of peptide therapeutics have heralded a profoundly promising paradigm shift. These innovative approaches capitalize on the intrinsic biological specificity of peptides, engineering them to precisely and selectively engage molecular targets predominantly expressed on malignant cells, thereby facilitating targeted therapeutic intervention while concomitantly minimizing collateral damage to the delicate surrounding healthy brain tissue. This comprehensive review endeavors to meticulously explore the current state-of-the-art in peptide-based therapies specifically tailored for pediatric brain tumors. It delves into their fundamental mechanisms of action, examines the breadth of their evolving clinical applications, and rigorously scrutinizes the complex scientific, technological, and logistical challenges that inherently accompany their rigorous development and eventual seamless implementation into routine clinical practice.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
1. Introduction
Pediatric brain tumors constitute an exceptionally heterogeneous category of intracranial neoplasms, distinguishing themselves as the most common solid tumors and the leading cause of cancer-related mortality in individuals under the age of 19. This diverse group encompasses a wide array of pathological entities, ranging from relatively benign, slow-growing lesions to highly aggressive, rapidly progressive malignancies. Among the most prevalent and therapeutically challenging are high-grade gliomas (HGGs), which include diffuse intrinsic pontine glioma (DIPG) – a particularly devastating form with a median survival measured in months – and medulloblastomas, the most common malignant brain tumor in children, which, despite advances, still carries significant morbidity and mortality, particularly for high-risk subgroups. Other significant types include ependymomas, craniopharyngiomas, and atypical teratoid/rhabdoid tumors (AT/RTs), each presenting unique diagnostic and therapeutic hurdles. Despite decades of intensive research and the application of maximally aggressive, multi-modality treatment regimens – often involving radical surgical resection where feasible, high-dose focal radiation therapy, and multi-agent systemic chemotherapy – the overall prognosis for children diagnosed with many of these aggressive tumors remains dishearteningly poor. For instance, the 5-year overall survival for children with DIPG hovers around 0-5%, a statistic that starkly underscores the profound urgency and critical imperative for the expeditious development of genuinely novel, more effective, and less toxic therapeutic strategies.
Traditional treatments, while often essential, are frequently limited by two primary factors: inherent resistance of tumor cells and unacceptable toxicity to the developing brain. The blood-brain barrier (BBB), a highly specialized and tightly regulated neurovascular unit, poses a formidable physiological obstacle, restricting the delivery of many conventional chemotherapeutic agents to the tumor site. Furthermore, the developing pediatric brain is exquisitely vulnerable to the cytotoxic effects of radiation and chemotherapy, leading to severe and often permanent long-term sequelae such as neurocognitive impairment, endocrine dysfunction, secondary malignancies, and vasculopathy, profoundly diminishing the quality of life for survivors. These critical limitations necessitate a paradigm shift towards therapies characterized by enhanced specificity and reduced systemic toxicity.
In this context, peptide therapeutics have unequivocally emerged as a profoundly promising and innovative modality, offering the tangible potential for highly targeted treatment with significantly reduced systemic toxicity. Peptides, being short chains of amino acids, possess several intrinsic advantages, including their high specificity for molecular targets, relatively straightforward and cost-effective synthesis, and generally favorable safety profiles. This comprehensive review endeavors to provide an in-depth and meticulously detailed analysis of the burgeoning role of peptide-based therapies within the complex and challenging landscape of pediatric brain tumors. It aims to meticulously highlight their diverse mechanisms of action, critically evaluate their expanding clinical applications, and systematically address the multifaceted scientific, engineering, and clinical challenges that are inevitably encountered throughout their arduous developmental trajectory, ultimately envisioning their transformative potential for this vulnerable patient population.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
2. Fundamental Mechanisms of Peptide-Based Therapies
Peptides are oligomers composed of amino acid residues linked by peptide bonds. Their inherent biological function is extraordinarily diverse, ranging from hormones and neurotransmitters to antimicrobial agents and immune modulators. In the context of targeted therapy, peptides are meticulously engineered to interact with high affinity and selectivity with specific molecular targets, such as cell surface receptors, intracellular enzymes, or protein-protein interaction domains, which are frequently overexpressed or uniquely dysregulated on the surface or within the cytoplasm of tumor cells, or within their supportive tumor microenvironment. Their relatively small size (typically 5 to 50 amino acids) confers several distinct advantages, including improved tissue penetration compared to larger antibody-based therapeutics, and the ability to be synthesized with high purity and consistency through established chemical or recombinant methods, making them highly attractive candidates for precision oncology. The principal mechanisms through which peptide therapeutics exert their profound therapeutic effects in cancer include:
2.1. Receptor-Mediated Targeting and Internalization
One of the most extensively exploited mechanisms involves the design of peptides that selectively bind to cell surface receptors that are either overexpressed or uniquely mutated on tumor cells compared to healthy cells. This differential expression provides a crucial therapeutic window. Upon binding, these peptides can act as agonists or antagonists, directly modulating receptor activity, or, more commonly, serve as molecular ‘homing devices’ to deliver cytotoxic payloads. The binding event can trigger receptor internalization, a process where the peptide-receptor complex is endocytosed into the cell, effectively delivering the attached therapeutic agent directly into the intracellular compartment of the tumor cell. This mechanism underpins several peptide-drug conjugate (PDC) strategies and peptide receptor radionuclide therapy (PRRT).
Key receptors frequently targeted in pediatric brain tumors and other cancers include:
- Somatostatin Receptors (SSTRs): Particularly SSTR2, SSTR3, and SSTR5, are often overexpressed in neuroendocrine tumors, including some pediatric brain tumors like certain gliomas and medulloblastomas. Peptides like octreotide and lanreotide, synthetic analogs of somatostatin, have been radiolabeled (e.g., with ^(111)In or ^(177)Lu) for both imaging and therapy (PRRT). These peptides are highly stable and resistant to enzymatic degradation, making them excellent candidates for in vivo applications [22, 23].
- Epidermal Growth Factor Receptor (EGFR) and its mutant variant EGFRvIII: EGFR is a receptor tyrosine kinase frequently amplified and/or mutated in many cancers, including high-grade gliomas. Peptides targeting EGFR can inhibit its downstream signaling pathways, crucial for cell proliferation and survival, or deliver cytotoxic agents. The EGFRvIII mutation, common in glioblastoma, presents a tumor-specific target, offering a unique opportunity for highly selective peptide-based therapies without affecting healthy tissues expressing wild-type EGFR.
- Integrins (e.g., αvβ3): These cell adhesion molecules are highly expressed on angiogenic endothelial cells and certain tumor cells, playing critical roles in cell migration, invasion, and angiogenesis. Peptides containing the RGD (Arginine-Glycine-Aspartic acid) motif specifically target integrins, making them useful for inhibiting angiogenesis, delivering drugs, or as imaging agents for tumor vasculature [16].
- Transferrin Receptor 1 (TfR1): Overexpressed on many rapidly proliferating tumor cells, including brain tumor cells, due to their increased iron demand. Peptides that mimic transferrin or bind to TfR1 can facilitate receptor-mediated transcytosis across the blood-brain barrier (BBB) and subsequent internalization into tumor cells, offering a promising strategy for targeted drug delivery to CNS malignancies [17].
- Gastrin-Releasing Peptide Receptors (GRPRs): Overexpressed in various cancers, including certain brain tumors. Bombesin and its analogs are GRPR-targeting peptides that have been explored for radionuclide imaging and therapy (e.g., Lutetium-177 NeoBOMB1, a bombesin analog, is under investigation for specific tumor types, though not exclusively pediatric brain tumors, it exemplifies the mechanism [12]).
2.2. Angiogenesis Inhibition
The growth and metastatic dissemination of solid tumors are critically dependent on the formation of new blood vessels, a process known as angiogenesis. Tumors secrete various pro-angiogenic factors that stimulate endothelial cell proliferation, migration, and new vessel formation, thereby supplying oxygen and nutrients essential for tumor sustenance. Peptides can be meticulously designed to interfere with this vital process, effectively ‘starving’ the tumor.
Mechanisms of angiogenesis inhibition by peptides include:
- Blocking Pro-angiogenic Growth Factors: Peptides can act as decoys or antagonists to prevent pro-angiogenic growth factors, such as Vascular Endothelial Growth Factor (VEGF) or Fibroblast Growth Factor (FGF), from binding to their respective receptors on endothelial cells. For example, certain synthetic peptides mimicking fragments of natural anti-angiogenic proteins (e.g., endostatin or angiostatin) can directly inhibit endothelial cell proliferation and migration. Pegdinetanib, while not solely a peptide, represents a fusion protein strategy where a peptide component contributes to its action, showing how peptide mimetics can target angiogenic pathways [10].
- Targeting Endothelial Cell Receptors: As mentioned, peptides containing the RGD motif can bind to integrins (e.g., αvβ3, αvβ5) highly expressed on activated endothelial cells in the tumor vasculature, thereby disrupting their signaling pathways critical for angiogenesis. This can lead to endothelial cell apoptosis and vessel regression [16].
- Disrupting Extracellular Matrix (ECM) Interactions: Some peptides can interfere with the interaction between endothelial cells and the ECM, which is crucial for vessel sprouting and maturation. By disrupting these interactions, peptides can destabilize the nascent tumor vasculature, making it more permeable and susceptible to other therapies, or leading to its regression.
2.3. Immune Modulation and Enhancement
The immune system plays a dual role in cancer: it can recognize and eliminate nascent tumor cells (immune surveillance), but tumors often develop sophisticated mechanisms to evade immune destruction. Peptides offer a powerful means to modulate the immune response, tipping the balance back in favor of anti-tumor immunity.
Strategies for immune modulation include:
- Peptide Vaccines: These vaccines utilize tumor-associated antigens (TAAs) or neoantigens derived from tumor cells, presented as short synthetic peptides. The goal is to prime and activate specific cytotoxic T lymphocytes (CTLs) that can recognize and destroy tumor cells expressing these antigens. The critical advantage is specificity, avoiding widespread immune activation. This mechanism is extensively discussed in Section 3.1 [20].
- Checkpoint Inhibition: While largely dominated by antibodies, the principles of immune checkpoint modulation can also be applied to peptides. Peptides could theoretically be designed to block inhibitory immune checkpoints (e.g., PD-1/PD-L1, CTLA-4) or activate co-stimulatory pathways, thereby unleashing or enhancing anti-tumor T cell responses. Research in this area is nascent for peptides but holds significant potential due to their smaller size and potential for improved tumor penetration [18].
- Antimicrobial Peptides (AMPs) with Immunomodulatory Roles: Beyond their direct cytotoxic effects, some AMPs have been shown to modulate immune responses. They can act as chemoattractants for immune cells, induce cytokine production, or influence antigen presentation. Their application in brain tumors might involve shaping the tumor microenvironment to be more pro-inflammatory and anti-tumorigenic.
- Oncolytic Peptides: These peptides can directly disrupt tumor cell membranes, leading to lysis and the release of tumor antigens, which can then be picked up by antigen-presenting cells (APCs) to initiate or amplify an anti-tumor immune response.
2.4. Direct Cytotoxicity and Apoptosis Induction
Certain peptides possess inherent cytotoxic properties, capable of directly inducing tumor cell death through various mechanisms. These are distinct from PDCs where the peptide primarily acts as a delivery vehicle for a separate cytotoxic agent.
- Membrane-Disrupting Peptides (Oncolytic Peptides): Many naturally occurring and engineered peptides, often cationic and amphipathic, can interact with and disrupt the negatively charged membranes of cancer cells, leading to pore formation and osmotic lysis. Their selectivity often arises from differences in membrane composition and charge between healthy and cancerous cells.
- Inhibition of Protein-Protein Interactions (PPIs): Protein-protein interactions are fundamental to almost all cellular processes, and their dysregulation is a hallmark of cancer. Peptides, particularly stapled peptides, can be engineered to mimic or disrupt critical PPIs involved in cell survival, proliferation, and apoptosis. A prime example is the p53-MDM2/MDMX pathway (discussed in 3.2), where peptides can reactivate the tumor suppressor p53 by disrupting its interaction with its negative regulators, MDM2 and MDMX, thereby initiating apoptosis [21].
- Enzyme Inhibition: Peptides can be designed as competitive inhibitors of enzymes that are essential for tumor growth and survival, such as proteases involved in metastasis or kinases involved in signaling pathways. Their high specificity can minimize off-target effects.
2.5. Diagnostic and Imaging Applications
Beyond therapy, peptides also serve as invaluable tools for diagnosis and imaging. By conjugating peptides with diagnostic radionuclides (e.g., ^(68)Ga, ^(99m)Tc, ^(18)F) or fluorescent dyes, they can be used to specifically visualize tumors and their metastases via PET (Positron Emission Tomography), SPECT (Single-Photon Emission Computed Tomography), or fluorescence imaging. This enables precise tumor localization, assessment of receptor expression levels, monitoring treatment response, and guiding surgical resection. The high specificity and rapid clearance of peptides contribute to excellent image contrast. For example, RGD peptides labeled with ^(18)F are used to image integrin expression, providing insights into tumor angiogenesis [1, 16].
Many thanks to our sponsor Esdebe who helped us prepare this research report.
3. Clinical Applications in Pediatric Brain Tumors
The application of peptide-based therapies in pediatric brain tumors represents a rapidly evolving field, driven by the critical need for more effective and less toxic treatment options. Several distinct peptide strategies are under intense investigation, each leveraging unique biological principles to target the vulnerabilities of these aggressive tumors.
3.1. Peptide Vaccines
Peptide vaccines represent an immunotherapeutic approach aimed at stimulating the host’s endogenous immune system to recognize and eradicate tumor cells. The fundamental premise is to introduce specific tumor-associated antigens (TAAs) or neoantigens – protein fragments that are either overexpressed, aberrantly expressed, or mutated in tumor cells but not in healthy tissues – to antigen-presenting cells (APCs), such as dendritic cells. These APCs then process and present the peptide antigens to T lymphocytes, particularly cytotoxic T lymphocytes (CTLs), in the context of Major Histocompatibility Complex (MHC) molecules. This presentation primes the T cells, leading to their activation, clonal expansion, and subsequent trafficking to the tumor site to specifically identify and lyse cancer cells expressing the target antigen [20].
In pediatric brain tumors, the challenge lies in identifying suitable tumor-specific antigens that are immunogenic and widely expressed across a significant proportion of tumor cells, while minimally expressed in normal brain tissue to avoid autoimmune neurotoxicity. Several promising targets have emerged:
- Cytomegalovirus (CMV) Antigens: A notable and extensively investigated example involves the peptide vaccine targeting antigens derived from Cytomegalovirus (CMV). CMV is a common herpesvirus that infects a large percentage of the population and often establishes latent infection. Intriguingly, CMV proteins, such as pp65, have been found to be expressed in a high percentage of various pediatric and adult high-grade gliomas, including glioblastoma, but are largely absent from adjacent normal brain tissue. This differential expression positions CMV antigens as highly attractive tumor-associated targets. Early-phase clinical trials, particularly at institutions like Duke University, have explored the safety and immunogenicity of CMV pp65 peptide vaccines in children with brain tumors, often in combination with other treatments. These trials have successfully demonstrated the vaccine’s ability to elicit a robust, CMV-specific T-cell immune response without significant systemic toxicity. Ongoing studies are now meticulously assessing the clinical efficacy of this approach, both as a monotherapy and in combination with standard-of-care treatments, aiming to prolong progression-free and overall survival (neurosurgery.duke.edu; Furtado et al., 2017 [1]).
- Mutant Histone H3 Peptides: Diffuse intrinsic pontine glioma (DIPG), a devastating pediatric brain tumor, is characterized by highly recurrent somatic mutations in histone H3 genes (e.g., H3K27M). These mutations lead to the production of neoantigens that are exclusively present in tumor cells. Developing peptide vaccines against these mutant histone H3 peptides represents a highly specific and promising strategy to target DIPG cells while sparing healthy tissue. Preclinical studies have shown that vaccination with H3K27M peptides can induce an anti-tumor immune response and improve survival in mouse models. Clinical trials are now being developed to translate these findings into human patients.
- WT1 (Wilms Tumor 1) Peptide: WT1 is an oncogenic transcription factor overexpressed in a variety of solid and hematological malignancies, including some pediatric brain tumors. Peptides derived from WT1 are being investigated in cancer vaccines due to their immunogenicity and widespread expression in tumor cells. Clinical trials in other cancers have shown promise, and its relevance to specific pediatric brain tumor subtypes is under active investigation.
- Survivin Peptides: Survivin is an inhibitor of apoptosis protein (IAP) that is overexpressed in most human cancers, including medulloblastomas and gliomas, and is associated with poor prognosis. Peptides derived from survivin can induce CTL responses against tumor cells expressing this protein. Its high expression in proliferating tumor cells and low expression in normal differentiated tissues makes it an attractive vaccine target.
- EGFRvIII Peptides: As mentioned in Section 2.1, the EGFRvIII mutation is highly tumor-specific. Peptide vaccines targeting this specific mutant epitope have been explored in adult glioblastoma and hold potential for pediatric gliomas harboring this mutation. This strategy aims for extremely high specificity, minimizing off-target effects.
The development of peptide vaccines faces challenges, including the need to overcome tumor-induced immune suppression, enhance vaccine immunogenicity (often requiring potent adjuvants or specific delivery systems like dendritic cell vaccines), and account for the diversity of MHC haplotypes in the patient population. However, their potential for highly specific, long-lasting anti-tumor immunity makes them a compelling area of research.
3.2. Stapled Peptides
One of the inherent limitations of linear therapeutic peptides is their conformational flexibility, which can lead to rapid enzymatic degradation in vivo and poor cell permeability. Stapled peptides represent an elegant solution to these challenges. This innovative class of peptides is chemically constrained to maintain a desired, often alpha-helical, secondary structure, which is crucial for their biological activity and ability to engage intracellular protein targets. The ‘staple’ is typically a hydrocarbon bridge formed through an intramolecular olefin metathesis reaction, though lactam and other chemical linkages are also used. This structural rigidity imparts several critical advantages [21]:
- Enhanced Stability: The staple significantly increases the peptide’s resistance to proteolytic degradation by peptidases and proteases, thereby extending its in vivo half-life.
- Improved Cell Permeability: The constrained conformation often facilitates efficient penetration of cell membranes, allowing stapled peptides to reach intracellular targets that are inaccessible to many conventional peptide and antibody therapeutics. This is particularly relevant for brain tumors where access to intracellular pathways is essential.
- Increased Target Affinity and Specificity: By locking the peptide into its bioactive conformation, stapling can enhance its binding affinity and specificity for its intended protein target, leading to more potent pharmacological effects.
Stapled peptides are particularly well-suited for disrupting critical protein-protein interactions (PPIs) that are often dysregulated in cancer cells. These PPIs frequently involve alpha-helical motifs, which the stapling technology can effectively mimic and stabilize. A prime example relevant to pediatric oncology is the targeting of the p53 tumor suppressor pathway:
- Targeting MDM2 and MDMX: The p53 tumor suppressor protein is a master regulator of cell cycle arrest, apoptosis, and DNA repair. In approximately half of human cancers, p53 itself is mutated. However, in cancers with wild-type p53 (including many pediatric brain tumors), its activity is often functionally suppressed by its negative regulators, MDM2 and MDMX. These proteins bind to the p53 transcription activation domain, preventing its function and promoting its degradation. Stapled peptides have been ingeniously designed to mimic the p53 alpha-helix that interacts with MDM2 and MDMX. By binding to the MDM2/MDMX pocket with high affinity, these stapled peptides effectively disrupt the p53-MDM2/MDMX interaction, thereby liberating and reactivating endogenous wild-type p53. Reactivated p53 then drives the transcription of its target genes, leading to cell cycle arrest and apoptosis specifically in tumor cells. Preclinical studies have shown highly promising results, demonstrating significant anti-tumor activity across a range of pediatric cancers, including neuroblastoma and medulloblastoma models (alexslemonade.org).
- Sulanemadlin (ALRN-6924): This is a prominent clinical-stage stapled peptide that functions as a dual inhibitor of MDM2 and MDMX. It has shown efficacy in preclinical models and is being investigated in clinical trials for adult and pediatric cancers with wild-type p53, including acute myeloid leukemia (AML) and peripheral T-cell lymphoma, demonstrating the clinical viability of this approach [11]. While direct pediatric brain tumor trials are still emerging, the foundational science indicates significant potential.
Further research is exploring stapled peptides that target other crucial PPIs in pediatric brain tumors, such as those involving the BCL-2 family proteins (which regulate apoptosis) or MYC family proteins (oncogenic transcription factors often dysregulated in medulloblastoma). The ability of stapled peptides to penetrate cells and modulate intracellular pathways makes them exceptionally attractive for addressing undruggable targets and overcoming intrinsic resistance mechanisms in pediatric brain tumors.
3.3. Peptide-Drug Conjugates (PDCs) and Peptide Receptor Radionuclide Therapy (PRRT)
Peptide-drug conjugates (PDCs) represent a powerful evolution in targeted therapy, combining the exquisite specificity and favorable pharmacokinetics of peptides with the potent cytotoxic efficacy of chemotherapy agents, toxins, or radionuclides. The fundamental design of a PDC involves three key components [24]:
- A Targeting Peptide: This component is specifically engineered to bind with high affinity to a receptor or antigen that is overexpressed on the surface of tumor cells or tumor vasculature (e.g., SSTRs, integrins, GRPRs, EGFR). The peptide acts as a ‘molecular key’ to unlock tumor cells.
- A Chemical Linker: This is a crucial, cleavable or non-cleavable bridge that connects the targeting peptide to the cytotoxic payload. Cleavable linkers are designed to release the payload specifically within the tumor microenvironment (e.g., pH-sensitive, enzyme-cleavable) or after internalization into the lysosomal compartment, maximizing intracellular drug concentration while minimizing systemic exposure. Non-cleavable linkers lead to the degradation of the entire conjugate within the cell, releasing active metabolites.
- A Cytotoxic Payload: This is the ‘warhead’ responsible for inducing cell death. Common payloads include highly potent small molecule chemotherapeutics (e.g., auristatins, maytansinoids, duocarmycins), bacterial toxins (e.g., diphtheria toxin, pseudomonas exotoxin), or radionuclides.
The primary advantage of PDCs is their ability to deliver a highly concentrated dose of a potent therapeutic agent directly to the tumor cells, thereby significantly enhancing efficacy and minimizing systemic toxicity to healthy tissues, a particularly vital consideration in pediatric patients. Upon binding to the target receptor, the PDC is typically internalized via receptor-mediated endocytosis, leading to the intracellular release of the cytotoxic payload.
Peptide Receptor Radionuclide Therapy (PRRT) is a specialized form of PDC therapy where the cytotoxic payload is a therapeutic radionuclide. This approach is particularly effective for tumors that strongly express specific cell surface receptors, allowing for the precise delivery of therapeutic radiation directly to tumor cells and their micrometastases. The emitted radiation (typically beta- or alpha-emitters) induces DNA damage, leading to tumor cell death [23].
Clinical applications and ongoing investigations in pediatric brain tumors include:
- Somatostatin Receptor (SSTR)-Targeting PDCs/PRRT: Many neuroendocrine tumors, including certain pediatric brain tumors like some types of gliomas and medulloblastomas, express somatostatin receptors (SSTRs), particularly SSTR2. Analogs of somatostatin, such as octreotide or octreotate, conjugated to radionuclides like Lutetium-177 (^177Lu-DOTATATE or ^177Lu-DOTATOC) or Yttrium-90 (^90Y-DOTATATE), are established treatments for neuroendocrine tumors in adults and are being explored for pediatric applications. These agents specifically bind to SSTRs on tumor cells, are internalized, and deliver localized radiation. This allows for both diagnostic imaging (e.g., with Gallium-68 DOTATATE PET scans) and targeted therapy. PRRT offers a systemic yet targeted radiation dose, potentially treating disseminated disease more effectively than external beam radiation, and has shown promise in some cases of refractory pediatric brain tumors and neuroblastomas [22, 23].
- GRPR-Targeting PDCs: Gastrin-Releasing Peptide Receptors (GRPRs) are frequently overexpressed in various human cancers, including prostate cancer, breast cancer, and some central nervous system tumors. Bombesin and its analogs are high-affinity ligands for GRPRs. Research into conjugating bombesin analogs with radionuclides (e.g., Lutetium-177 NeoBOMB1 [12]) or chemotherapy drugs is underway to target GRPR-positive pediatric brain tumors. This strategy holds potential for both diagnostic imaging and targeted radiotherapy.
- Integrin-Targeting PDCs: Peptides containing the RGD motif can be conjugated to various payloads (chemotherapeutics, toxins, or radionuclides) to target integrins overexpressed on tumor endothelial cells and certain tumor cells. This approach can simultaneously inhibit angiogenesis and deliver a cytotoxic punch to both tumor cells and their supporting vasculature. While not yet in widespread clinical use for pediatric brain tumors, preclinical data support its potential [16].
- Targeting of other overexpressed receptors: Research continues to identify and exploit other tumor-specific or tumor-associated receptors for PDC development, aiming to deliver highly potent agents with enhanced precision. The challenge remains to find receptors with sufficiently high and selective expression in pediatric brain tumors to ensure a favorable therapeutic index.
3.4. Other Emerging Peptide-Based Strategies
Beyond the more established categories, several other innovative peptide-based strategies are garnering attention for their potential in pediatric brain tumor therapy:
- Cell-Penetrating Peptides (CPPs): While not inherently therapeutic, CPPs are short peptides that can facilitate the intracellular delivery of various cargoes (e.g., small molecules, nucleic acids, larger peptides, nanoparticles) across biological membranes, including potentially the BBB. By conjugating a therapeutic peptide or another drug to a CPP, researchers aim to improve its brain penetrance and cellular uptake, thereby enhancing its therapeutic efficacy for intracellular targets.
- Modulators of the Tumor Microenvironment: The tumor microenvironment (TME) is a complex ecosystem of cells (fibroblasts, immune cells, endothelial cells), extracellular matrix (ECM), and soluble factors that profoundly influences tumor growth, invasion, and response to therapy. Peptides can be designed to target specific components of the TME, such as matrix metalloproteinases (MMPs) involved in ECM remodeling, or growth factors and cytokines that promote tumor progression. By normalizing or disrupting the TME, peptides can render tumors more susceptible to conventional treatments or directly inhibit their growth.
- Peptide-Based PROTACs (Proteolysis Targeting Chimeras): PROTACs are bifunctional molecules that induce the degradation of specific target proteins by recruiting them to an E3 ubiquitin ligase. While small molecule PROTACs are more common, stapled peptides or other engineered peptides can be utilized as the ‘ligand’ component, particularly for difficult-to-drug protein targets that are crucial for tumor survival. For example, the reference to ‘Stapled Peptide PROTAC’ [4] highlights this emerging area, suggesting its potential for pediatric solid tumors by inducing targeted protein degradation rather than just inhibition.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
4. Intricate Challenges in Development and Implementation
Despite the remarkable promise inherent in peptide therapeutics, their successful translation from preclinical studies to widespread clinical application in pediatric brain tumors is fraught with a series of intricate scientific, technological, and logistical challenges. Addressing these hurdles necessitates innovative solutions and concerted multidisciplinary efforts.
4.1. Delivery and in vivo Stability: Navigating the Blood-Brain Barrier
The most formidable barrier to effective CNS drug delivery, particularly for peptide therapeutics, is the blood-brain barrier (BBB). This highly specialized and tightly regulated neurovascular structure is composed of endothelial cells interconnected by tight junctions, surrounded by pericytes and astrocytic end-feet, creating a physical and metabolic barrier that strictly controls the passage of substances from the bloodstream into the brain parenchyma. While essential for protecting the delicate CNS from toxins and pathogens, the BBB represents a major impediment for most therapeutic agents, including many peptides, which often have unfavorable physicochemical properties (e.g., relatively large size, hydrophilicity, susceptibility to efflux pumps) for passive diffusion across this barrier [13, 17, 19].
Key challenges and potential solutions include:
-
Blood-Brain Barrier Penetration: Most peptides are too large and hydrophilic to passively diffuse across the BBB. Strategies to circumvent or bypass the BBB include:
- Receptor-Mediated Transcytosis (RMT): Exploiting endogenous BBB receptors that naturally transport essential molecules into the brain (e.g., transferrin receptor, insulin receptor). Peptides can be engineered to bind to these receptors, initiating their internalization and transcytosis across the endothelial cells into the brain. For instance, peptides derived from transferrin or antibodies targeting TfR1 can carry payloads across the BBB [17].
- Adsorption-Mediated Transcytosis (AMT): Utilizing positively charged peptides or nanoparticles to interact with negatively charged proteoglycans on the luminal surface of endothelial cells, leading to non-specific adsorption and subsequent transcytosis.
- Efflux Pump Inhibition: The BBB expresses efflux transporters (e.g., P-glycoprotein, BCRP) that actively pump out many xenobiotics, including drugs, from the brain. Co-administration with efflux pump inhibitors or designing peptides resistant to efflux can enhance brain penetration [17].
- Disruption of the BBB: While historically explored (e.g., mannitol-induced osmotic disruption, focused ultrasound), generalized BBB disruption carries risks of increased permeability to unwanted substances and inflammation, making it a less favored approach for routine pediatric use.
- Intrathecal or Intracerebroventricular Administration: Direct administration into the cerebrospinal fluid (CSF) bypasses the BBB, ensuring local drug delivery to the CNS. This approach is invasive and limits drug distribution to superficial brain regions, but can be effective for certain indications, particularly leptomeningeal disease.
- Convection-Enhanced Delivery (CED): Involves continuous, positive-pressure infusion of therapeutic agents directly into the brain parenchyma via implanted catheters. This method bypasses the BBB and allows for more uniform and widespread distribution within a defined region, but is highly invasive.
-
In vivo Stability and Pharmacokinetics: Peptides are inherently susceptible to rapid enzymatic degradation by circulating proteases (peptidases) and endopeptidases, leading to very short in vivo half-lives (often minutes). This rapid clearance necessitates frequent dosing or high concentrations, which can be impractical or lead to systemic toxicity. Strategies to enhance stability and improve pharmacokinetics include [14, 15]:
- Amino Acid Modifications: Incorporating D-amino acids, unnatural amino acids, or N-methylation can increase resistance to enzymatic cleavage.
- Cyclization: Forming cyclic peptides can restrict conformational flexibility, making them less susceptible to proteolysis and improving binding affinity.
- Stapling: As discussed, hydrocarbon stapling significantly enhances stability and cell permeability [21].
- PEGylation: Covalent attachment of polyethylene glycol (PEG) chains to peptides increases their hydrodynamic radius, reducing renal clearance and masking them from enzymatic degradation and immune recognition, thereby extending their circulating half-life.
- Albumin Binding: Engineering peptides to reversibly bind to serum albumin can also prolong their half-life by reducing glomerular filtration.
- Formulation Strategies: Encapsulation in nanoparticles (e.g., liposomes, polymeric nanoparticles) can protect peptides from degradation, facilitate BBB penetration, and enable sustained release.
4.2. Immunogenicity and Allergic Reactions
As exogenous biomolecules, peptides, especially those derived from non-human sources or with modifications, can be recognized as foreign by the host immune system. This immunogenicity can trigger an immune response, leading to the production of anti-drug antibodies (ADAs). ADAs can have several detrimental effects:
- Reduced Efficacy: By binding to the therapeutic peptide, ADAs can neutralize its activity, prevent its interaction with its target, or accelerate its clearance from circulation, thereby reducing its therapeutic efficacy.
- Altered Pharmacokinetics: The formation of immune complexes can alter the peptide’s distribution, metabolism, and excretion, leading to unpredictable drug exposure.
- Allergic/Hypersensitivity Reactions: In some cases, the immune response can manifest as mild to severe allergic reactions, including anaphylaxis, posing significant safety concerns for patients.
Strategies to mitigate immunogenicity include:
- Humanization/De-immunization: For peptides derived from non-human sequences, modifying them to resemble human sequences can reduce immunogenicity. Computational tools can predict immunogenic epitopes, allowing for their modification.
- Conformational Masking: Techniques like PEGylation can mask potential immunogenic epitopes from immune recognition.
- Tolerance Induction: In some experimental settings, strategies to induce immune tolerance to the peptide are explored, though this is complex and not routinely applied.
4.3. Manufacturing Scalability and Cost-Effectiveness
The production of clinical-grade peptides, particularly for widespread use, presents significant manufacturing challenges and economic considerations:
- Complex Synthesis: Peptide synthesis, especially for longer or modified peptides (e.g., stapled, cyclized, or those with unnatural amino acids), can be complex and expensive. Solid-phase peptide synthesis (SPPS) is the dominant method for short-to-medium length peptides, but requires specialized reagents, significant purification steps, and can be limited in scale. Liquid-phase peptide synthesis (LPPS) or recombinant production in bacterial or eukaryotic systems are alternatives for larger scales or specific peptide types, but each has its own complexities and limitations [15].
- Purity Requirements: Therapeutic peptides demand extremely high purity (typically >95-98%) to ensure safety and efficacy, requiring extensive and costly purification processes (e.g., HPLC). Impurities can be immunogenic or toxic.
- Quality Control: Stringent quality control measures are essential at every stage of manufacturing to ensure batch-to-batch consistency, identity, purity, and potency, adding to the overall cost and complexity.
- Regulatory Hurdles: Obtaining regulatory approval for novel peptide therapeutics, particularly those with complex modifications or delivery systems, involves rigorous preclinical testing and multiple phases of clinical trials, which are time-consuming and expensive.
These factors collectively contribute to the high cost of peptide therapeutics, which can limit their accessibility and affordability, particularly for chronic conditions or in healthcare systems with constrained resources. Innovations in manufacturing technology, such as flow chemistry and enzymatic synthesis, are continuously sought to improve efficiency and reduce costs.
4.4. Tumor Heterogeneity and Acquired Resistance
Pediatric brain tumors, like many cancers, exhibit significant intratumoral and intertumoral heterogeneity, meaning that different cells within the same tumor or tumors of the same diagnostic type can have diverse molecular profiles. This heterogeneity poses a challenge for targeted therapies, as some tumor cell clones may not express the target receptor or pathway, leading to inherent resistance. Furthermore, tumors can evolve under therapeutic pressure, developing acquired resistance mechanisms, such as:
- Downregulation or Mutation of the Target Receptor: Tumor cells can reduce the expression of the targeted receptor or acquire mutations that prevent peptide binding, thereby evading therapy.
- Activation of Bypass Signaling Pathways: Tumors can activate alternative signaling pathways to compensate for the inhibited target pathway, maintaining their proliferative and survival advantages.
- Increased Drug Efflux: Upregulation of efflux pumps can lead to the removal of peptide-drug conjugates from the cells.
Addressing heterogeneity and resistance often requires combination therapies targeting multiple pathways, or sequential therapies guided by dynamic monitoring of tumor molecular profiles.
4.5. Off-Target Effects and Toxicity
While peptides are designed for high specificity, perfect selectivity is rarely achieved. Some degree of off-target binding to receptors or proteins expressed in healthy tissues can occur, leading to unintended side effects. Even if a target is overexpressed in tumors, low-level expression in critical normal organs could lead to toxicity. For example, if a peptide targets a receptor expressed in both tumor and kidney, nephrotoxicity could be a concern. Meticulous preclinical toxicology studies and careful dose escalation in clinical trials are essential to identify and manage these potential off-target effects, ensuring a favorable therapeutic index, especially in vulnerable pediatric populations.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
5. Future Directions and Synergistic Opportunities
The field of peptide therapeutics for pediatric brain tumors is poised for significant advancements, driven by continuous innovation in peptide engineering, drug delivery systems, and a deeper understanding of tumor biology. Future directions are focused on overcoming current limitations and exploring synergistic approaches to maximize therapeutic efficacy and minimize toxicity.
5.1. Advanced Peptide Engineering and Design
- Computational Peptide Design: In silico methods, including molecular docking, molecular dynamics simulations, and machine learning algorithms, are increasingly being used to predict peptide-target interactions, design novel peptides with enhanced affinity and specificity, and optimize properties like stability and cell permeability. This accelerates the discovery and optimization process.
- Macrocyclization and Constrained Peptides: Beyond stapling, other methods of macrocyclization (e.g., disulfide bridges, head-to-tail cyclization) are being explored to further rigidify peptides, improve their stability, and enhance their ability to bind to challenging targets, including intracellular PPIs [21].
- De novo Peptide Design: Moving beyond peptides derived from natural ligands, researchers are designing entirely new peptide sequences from scratch, guided by structural biology and computational modeling, to target specific binding pockets or functional sites on proteins.
- Bifunctional and Multifunctional Peptides: Designing peptides that can engage multiple targets simultaneously (e.g., a peptide that targets a tumor receptor and also binds to an immune cell receptor to activate an anti-tumor response) or perform multiple functions (e.g., targeting and crossing the BBB, then delivering a drug, and then being used for imaging). This can lead to synergistic effects and overcome resistance.
5.2. Integration with Nanotechnology for Enhanced Delivery
Nanotechnology offers a transformative platform for improving the delivery, stability, and bioavailability of peptide therapeutics, particularly across the BBB.
- Nanoparticle Encapsulation: Encapsulating peptides within various nanoparticles (e.g., liposomes, polymeric nanoparticles, solid lipid nanoparticles, dendrimers) can shield them from enzymatic degradation, prolong their circulation time, and facilitate their transport across the BBB. Nanoparticles can be surface-functionalized with targeting ligands (e.g., transferrin, specific peptides) to achieve active targeting to brain tumor cells or BBB receptors [17].
- BBB-Penetrating Nanocarriers: Engineered nanocarriers can leverage receptor-mediated transcytosis pathways or physical disruption methods (e.g., focused ultrasound with microbubbles) to deliver peptide payloads specifically to the brain tumor microenvironment.
- Theranostic Nanoparticles: Combining therapeutic peptides with diagnostic imaging agents (e.g., fluorescent dyes, MRI contrast agents, radionuclides) within a single nanocarrier allows for simultaneous targeted therapy and real-time monitoring of drug delivery, tumor response, and disease progression.
5.3. Combination Therapies and Immunotherapy Synergy
Recognizing the complexity and heterogeneity of pediatric brain tumors, future therapeutic strategies will increasingly focus on rational combination approaches, where peptide-based therapies are integrated with other modalities to achieve synergistic effects and overcome resistance.
- Peptides with Immunotherapy: Combining peptide vaccines with immune checkpoint inhibitors (e.g., PD-1/PD-L1 antibodies) could potentially unleash a more potent and durable anti-tumor immune response [18]. Peptide-based approaches could also be combined with CAR T-cell therapies (e.g., B7-H3 CAR T-cell therapy, which has received Breakthrough Therapy Designation for pediatric brain tumors [7, 8]), perhaps by targeting different antigens or modulating the tumor microenvironment to enhance CAR T-cell efficacy. (seattlechildrens.org).
- Peptides with Radiation Therapy: PRRT is an inherent combination, but peptides can also sensitize tumor cells to external beam radiation therapy, enhancing the efficacy of standard radiation protocols while potentially allowing for reduced radiation doses and thus fewer long-term side effects.
- Peptides with Chemotherapy/Targeted Small Molecules: PDCs are a form of combination, but peptides could also be used to target and deliver specific small molecule inhibitors or traditional chemotherapeutics that otherwise struggle with BBB penetration or systemic toxicity. For example, peptides could enhance the delivery of MEK inhibitors like Selumetinib [9] if it were a payload in a PDC, leading to improved efficacy.
- Multi-Modal Approaches: The integration of surgery, radiation, chemotherapy, targeted therapies, and peptide therapeutics in a precisely sequenced and personalized manner offers the best hope for durable responses in complex pediatric brain tumors.
5.4. Personalized Medicine and Biomarker-Driven Trials
The increasing understanding of the molecular landscape of pediatric brain tumors (e.g., through genomic sequencing) allows for the development of personalized medicine approaches. Future peptide therapeutic development will increasingly be guided by robust biomarkers:
- Molecular Profiling: Routine molecular profiling of individual patient tumors will identify specific receptor overexpression, mutations (e.g., H3K27M), or signaling pathway dysregulations that can be specifically targeted by peptide therapies. This ensures that the right peptide is given to the right patient.
- Response Biomarkers: Developing predictive and prognostic biomarkers to identify patients most likely to respond to a particular peptide therapy, as well as companion diagnostics to monitor treatment efficacy and detect early signs of resistance.
- Organoid and Patient-Derived Xenograft (PDX) Models: Utilizing patient-derived organoids and PDX models allows for personalized ex vivo and in vivo testing of peptide therapies, identifying the most effective treatments for an individual patient’s tumor and guiding clinical trial design.
5.5. Collaborative Initiatives and Regulatory Support
The complex nature of pediatric brain tumor research necessitates robust collaborative efforts. Organizations like the Pediatric Brain Tumor Consortium (PBTC) and the Pediatric Early Phase Clinical Trials Network (PEP-CTN) are absolutely vital in accelerating the translation of innovative therapies from basic science discoveries to clinical trials and ultimately to patient care (dctd.cancer.gov; cancer.gov). These consortia facilitate multi-institutional trials, standardize protocols, and pool resources, which are crucial for rare diseases like pediatric brain tumors. Furthermore, regulatory bodies (e.g., FDA) are increasingly offering expedited review pathways (e.g., Breakthrough Therapy Designation, Regenerative Medicine Advanced Therapy Designation, as seen with B7-H3 CAR T-cell therapy [7, 8]) for promising therapies addressing serious unmet medical needs in pediatric populations, which can significantly shorten the development timeline for peptide therapeutics.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
6. Conclusion
Pediatric brain tumors continue to represent a devastating diagnosis for countless families, demanding relentless pursuit of more effective and less toxic therapeutic paradigms. The landscape of traditional treatments, while foundational, is inherently limited by concerns of efficacy, systemic toxicity to the developing CNS, and the formidable physiological barrier presented by the blood-brain barrier. In this critical context, peptide therapeutics have unequivocally emerged as a highly novel and profoundly promising approach to the treatment of these challenging malignancies. Their intrinsic advantages, including exceptional specificity, generally favorable safety profiles, and amenability to sophisticated engineering, position them as potential game-changers in pediatric neuro-oncology.
This review has underscored the diverse and sophisticated mechanisms by which peptides exert their anti-tumor effects, ranging from highly specific receptor-mediated targeting and the precise delivery of cytotoxic payloads via peptide-drug conjugates and radionuclide therapies, to the intricate modulation of tumor angiogenesis and the potent activation of anti-tumor immune responses through peptide vaccines and immunomodulatory agents. The advancements in technologies such as stapled peptides have further expanded their therapeutic reach, enabling the targeting of previously ‘undruggable’ intracellular protein-protein interactions essential for tumor survival.
While the journey towards widespread clinical implementation is not without its significant challenges – particularly in overcoming the formidable hurdles of ensuring adequate brain delivery, enhancing in vivo stability, mitigating immunogenicity, and addressing the complexities of manufacturing and cost – ongoing research continues to push the boundaries of our understanding and application of these sophisticated therapies. Future directions emphasize the critical role of advanced peptide engineering, the transformative potential of nanotechnology for enhanced delivery across the blood-brain barrier, and the imperative for rational combination therapies that synergize peptide-based approaches with other established or emerging modalities like immunotherapy and targeted small molecules. Furthermore, the increasing adoption of personalized medicine strategies, guided by comprehensive molecular profiling and robust biomarkers, will be pivotal in tailoring these treatments to individual patients, maximizing therapeutic benefit while minimizing adverse effects.
The collaborative efforts of academic institutions, industry partners, patient advocacy groups, and regulatory bodies, exemplified by consortia like the Pediatric Brain Tumor Consortium, are absolutely essential for accelerating the translation of these innovative therapies from fundamental bench research to the patient’s bedside. With sustained innovation, rigorous preclinical and clinical investigation, and unwavering collaboration, peptide-based treatments hold the profound potential to offer a significantly more effective and remarkably less toxic alternative or complement to current standard-of-care therapies, ultimately transforming the prognosis and improving the quality of life for children afflicted with brain tumors.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
References
- Furtado AD, Ceschin R, Blüml S, et al. Neuroimaging of Peptide-based Vaccine Therapy in Pediatric Brain Tumors: Initial Experience. Neuroimaging Clin N Am. 2017 Feb;27(1):155-166. (cancer.ucsf.edu)
- Thompson EM, et al. Duke Neurosurgery granted funding to study novel vaccine for pediatric brain tumors. Duke Department of Neurosurgery. October 4, 2021. (neurosurgery.duke.edu)
- Pediatric Brain Tumor Consortium (PBTC) and Pediatric Early Phase Clinical Trials Network (PEP-CTN). National Cancer Institute. (dctd.cancer.gov)
- Stapled Peptide PROTAC: A 3-in-1 Treatment for Pediatric Solid Tumors. Alex’s Lemonade Stand Foundation for Childhood Cancer. (alexslemonade.org)
- Seattle Children’s Launches Pioneering Immunotherapy Clinical Trial. Seattle Children’s. April 25, 2023. (seattlechildrens.org)
- Clinical Trials to Treat Childhood Embryonal Tumors. National Cancer Institute. (cancer.gov)
- FDA Grants Breakthrough Therapy Designation for BrainChild Bio’s B7-H3 CAR T-Cell Therapy for Incurable Pediatric Brain Tumors. Business Wire. April 22, 2025. (businesswire.com)
- FDA Grants Regenerative Medicine Advanced Therapy Designation for BrainChild Bio’s B7-H3 CAR T-cell Therapy for Incurable Pediatric Brain Tumors. Business Wire. May 15, 2025. (businesswire.com)
- Selumetinib. Wikipedia. (en.wikipedia.org)
- Pegdinetanib. Wikipedia. (en.wikipedia.org)
- Sulanemadlin. Wikipedia. (en.wikipedia.org)
- Lutetium-177 NeoBOMB1. Wikipedia. (en.wikipedia.org)
- Barboza R, de Oliveira E, de Araujo R. The challenges of brain drug delivery. Braz J Med Biol Res. 2018;51(10):e7805.
- Lau JL, Dunn MK. Therapeutic Peptides: Historical Perspectives, Current Trends, and Future Directions. J Pharm Sci. 2018 Sep;107(9):1974-1981.
- Vlieghe P, Lisowski L, Martinez J, Khrestchatisky M. Synthetic therapeutic peptides: science and market. Drug Discov Today. 2010 Sep;15(17-18):40-56.
- Horton L, et al. Peptide-based therapeutics for cancer: recent advances and future prospects. Int J Pept Res Ther. 2020;26(1):185-201.
- Ametzazurra A, et al. Brain tumors, the blood-brain barrier and strategies for drug delivery. Front Pharmacol. 2021;12:690559.
- Hodi FS, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med. 22 Aug 2010;363(8):711-23. (General reference for immune checkpoint inhibitors, not peptide specific, but relevant to combination therapy section).
- Glickson JD. Targeted drug delivery to brain tumors. Discov Med. 2009 Aug;8(43):64-70.
- Walczak M, et al. Peptide vaccines: a promising approach against cancer. OncoTargets Ther. 2017 Apr;10:2039-2049.
- Chang K, et al. Stapled Peptides in Drug Discovery: Recent Advances and Future Outlook. Molecules. 2018 Dec 20;23(12):3361.
- Reubi JC. Peptide receptors as molecular targets for cancer diagnosis and therapy. Endocr Rev. 2003 Aug;24(4):389-421.
- Zitzmann S, et al. Targeted radionuclide therapy for neuroblastoma: A systematic review. Int J Mol Sci. 2022 Jul 26;23(15):8249.
- Munteanu M, et al. Peptide-based drug conjugates for cancer therapy: A review. Int J Mol Sci. 2021 Oct 22;22(21):11417.

Be the first to comment