The investigational agent CyPep-1, now predominantly referred to as CY-101 in its oncological development by Cytovation AS, represents a novel, first-in-class therapeutic candidate. It is a synthetic, 27 D-amino acid cationic lytic peptide engineered for the treatment of various cancers.[1] A significant attribute of CY-101 is its innovative dual mechanism of action. The peptide directly targets and disrupts cancer cell membranes, leading to rapid cell lysis and the release of tumor antigens, a process designed to induce immunogenic cell death and an in situ vaccination effect.[3] Concurrently, CY-101 has been shown to inhibit the Wnt/β-catenin signaling pathway, a critical oncogenic driver in many malignancies.[2] This dual approach distinguishes CY-101 from many other oncolytic agents. The evolution in nomenclature from the general research term "CyPep-1" to the oncology-specific "CY-101" signifies a deliberate strategic focusing of its development program, reflecting an increasing understanding of its specific mechanisms in cancer and a commitment to this therapeutic area. This is further differentiated from an earlier formulation, CyPep-H1, which was explored for dermatological conditions.[7] Such shifts in designation often occur as a product matures and its lead indication becomes more clearly defined, underscoring Cytovation's strategic direction for CY-101 in oncology.
Preclinical studies have demonstrated significant efficacy for CY-101 across a range of cancer models, including breast cancer, osteosarcoma, glioma, and, notably, Adrenocortical Carcinoma (ACC), colorectal cancer (CRC), and melanoma.[1] These studies have highlighted its selectivity for cancer cells over normal cells and its ability to work synergistically with checkpoint inhibitors like anti-PD-1 agents.[3] The combination of direct oncolysis and Wnt/β-catenin pathway modulation offers a unique therapeutic strategy, potentially overcoming resistance mechanisms associated with single-pathway targeting and broadening its applicability, especially in Wnt-driven tumors.[2]
The clinical development of CY-101 is centered around the Phase I/IIa CICILIA trial (NCT04260529), which is evaluating the safety, tolerability, pharmacokinetics, and preliminary efficacy of intratumorally administered CY-101, both as a monotherapy and in combination with pembrolizumab, in patients with advanced solid tumors.[3] Early results from this trial have been promising, indicating a favorable safety profile with no dose-limiting toxicities observed in the monotherapy dose-escalation phase, and establishing a recommended Phase 2 dose (RP2D) of 20 mg.[2] Importantly, early signs of clinical efficacy have been observed, particularly in patients with ACC whose tumors exhibit dysregulated Wnt/β-catenin signaling.[2] This empirical evidence, targeting a rare disease with high unmet medical need, has led to a strategic prioritization of ACC.
Reflecting this focus and the promising data, CY-101 has received Orphan Drug Designation from the U.S. Food and Drug Administration (FDA) for the treatment of ACC.[2] This regulatory milestone, coupled with international collaborations with entities like Cancer Research UK and the Norwegian Cancer Society for a dedicated Phase II ACC trial [6], underscores a "fast-to-market" strategy. ACC often has limited effective treatment options, making it an area where a novel agent like CY-101 can make a significant impact.
Overall, CY-101 (CyPep-1) is emerging as a promising novel immunotherapy. Its dual mechanism of action, favorable early clinical safety, and encouraging efficacy signals, especially in difficult-to-treat Wnt-driven cancers like ACC, position it as a candidate with significant therapeutic potential for patients with solid tumors.
The landscape of cancer therapy continually seeks innovative approaches to address the challenges posed by advanced solid tumors, treatment resistance, and the need for therapies with improved efficacy and safety profiles. Lytic peptides have emerged as a promising class of therapeutic agents due to their potential to directly kill cancer cells and stimulate an anti-tumor immune response.[1] These peptides often exploit fundamental biophysical differences between cancer cell membranes and those of normal cells, such as surface charge and fluidity. CyPep-1 was conceived from this rationale, designed as a novel cationic peptide composed of 27 D-amino acids, with the primary goal of selectively targeting and lysing cancer cells, thereby offering a new modality in the fight against various malignancies.[1] The development aimed to harness the direct cytotoxic potential of membrane disruption while potentially minimizing damage to healthy tissues and engaging the host immune system for a more durable anti-cancer effect.[3]
To ensure clarity throughout this report, it is important to address the nomenclature associated with this investigational agent:
The transition from a university-developed peptide to the lead asset of a dedicated biotechnology company, Cytovation AS, accompanied by significant international collaborations, points to a strong foundational science and a widely perceived clinical potential. This evolution is common in pharmaceutical development, where initial discoveries are translated into focused therapeutic programs.
Cytovation AS is a clinical-stage immune-oncology company headquartered in Bergen, Norway. The company originated as a spin-off from the University of Bergen and is dedicated to the development of CY-101 (CyPep-1) as its lead therapeutic candidate.[2] Cytovation has strategically entered into key collaborations to advance the clinical development of CY-101, leveraging external expertise and resources:
The ability of Cytovation to attract such high-caliber partners underscores the external validation of CY-101's technology and the strategic, data-driven approach being taken to its clinical development. These collaborations are instrumental in de-risking the development process, providing financial and operational leverage, and accelerating the journey of CY-101 towards potentially benefiting patients.
CY-101 (CyPep-1) is a synthetic cationic lytic peptide meticulously designed for therapeutic application. It is composed of a specific sequence of 27 D-amino acids: Tyr-Gly-Arg-Lys-Lys-Arg-Arg-Gln-Arg-Arg-Arg-Gly-Lys-Thr-Leu-Arg-Val-Ala-Lys-Ala-Ile-Tyr-Lys-Arg-Tyr-Ile-Glu (abbreviated as YGRKKRRQRRRGKTLRVAKAIYKRYIE).[9] This D-amino acid composition is a critical design feature, as it confers significant stability against proteolytic degradation by endogenous enzymes, a common challenge for therapeutic peptides made of naturally occurring L-amino acids.[1] This enhanced stability is crucial for maintaining its structural integrity and biological activity within the tumor microenvironment following administration.
The peptide has a calculated molecular weight of 3451.09 Da and a high theoretical isoelectric point (pI) of 11.81, reflecting its strong cationic (positively charged) nature at physiological pH.[1] This positive charge is fundamental to its initial interaction with the generally negatively charged surfaces of cancer cells. For clinical application in oncology, CY-101 is administered directly into the tumor via intratumoral injection.[3] The stability afforded by its D-amino acid structure is an uncommon but highly advantageous feature for a peptide therapeutic, potentially contributing to its sustained local activity and observed potency.[1]
Table 1: CyPep-1 / CY-101 - Key Drug Characteristics
Feature | Description | Source(s) |
---|---|---|
Nomenclature | CyPep-1 (original research peptide), CY-101 (oncology development) | 2 |
Developer | Cytovation AS | 6 |
Peptide Type | Cationic Lytic Peptide | 1 |
Key Structural Features | 27 D-Amino Acids; Sequence: YGRKKRRQRRRGKTLRVAKAIYKRYIE | 1 |
Molecular Weight | 3451.09 Da | 9 |
Theoretical pI | 11.81 | 1 |
Primary Mechanism | Targeted Cell Membrane Lysis & Immunogenic Cell Death | 1 |
Secondary Mechanism (CY-101) | Wnt/β-Catenin Pathway Inhibition | 2 |
Administration Route | Intratumoral Injection | 3 |
Key Stability Feature | D-amino acid composition confers resistance to proteolytic degradation | 1 |
The principal and initially characterized mechanism of action for CY-101 is its potent and selective membranolytic activity against cancer cells.[1] This activity stems from the peptide's strong cationic nature, which facilitates electrostatic interactions with the predominantly negatively charged outer surfaces of cancer cell membranes.[1] Cancer cells frequently exhibit an altered membrane composition, including an increased exposure of anionic phospholipids like phosphatidylserine (PS) on their outer leaflet, which provides a degree of selectivity for cationic peptides like CY-101 over normal cells that typically maintain a more neutral outer membrane surface.[1]
Upon binding to the cancer cell membrane, CY-101 is believed to disrupt its integrity by forming pores or channels.[1] This process, visually confirmed by scanning electron microscopy (SEM) showing physical holes and time-lapse confocal microscopy demonstrating leakage of cellular contents [1], leads to a rapid loss of osmotic balance, membrane depolarization, and ultimately, cell lysis. The proposed models for this membrane disruption include the "toroidal pore" or "wormhole model," where peptides and lipids together form the pore lining, or the "carpet model," where peptides accumulate on the membrane surface, causing tension and eventual permeabilization.[1]
A critical consequence of this direct cell lysis is the release of a broad array of intracellular contents, including tumor-specific antigens (TSAs) and damage-associated molecular patterns (DAMPs), into the tumor microenvironment (TME).[2] This release is highly significant from an immunological perspective, as it can transform the tumor site into a hub of immune stimulation. The liberated TSAs can be taken up by antigen-presenting cells (APCs), such as dendritic cells, which then process and present these antigens to T cells, initiating a tumor-specific adaptive immune response. The DAMPs concurrently promote an inflammatory microenvironment, further enhancing APC maturation and immune cell recruitment. This entire process is often referred to as an in situ vaccination, where the dying tumor cells themselves provide the antigenic material to prime a systemic, targeted anti-tumor immunity.[2]
Beyond its direct lytic effects, CY-101 has been identified as possessing a crucial secondary mechanism of action: the inhibition of the Wnt/β-catenin signaling pathway.[2] This pathway is a fundamental cellular signaling cascade that, when aberrantly activated, plays a significant role in the development and progression of numerous cancers by promoting cell proliferation, survival, stemness, and by modulating the tumor microenvironment to be more immunosuppressive.[24]
CY-101 is described as a "membranolytic inhibitor" of this pathway, suggesting a potential link between its membrane interaction and its ability to modulate intracellular signaling.[6] One proposed mechanism for this inhibition involves the activation of Axin2 [2], a key component of the β-catenin destruction complex whose upregulation typically leads to increased β-catenin degradation, thereby reducing Wnt signaling output. By inhibiting the oncogenic Wnt/β-catenin pathway, CY-101 aims to achieve several anti-tumor effects:
The dysregulation of the Wnt/β-catenin pathway is a known characteristic of a wide range of cancers, including colorectal cancer, hepatocellular carcinoma, some forms of lung and ovarian cancer, and notably, is a very frequent driver in Adrenocortical Carcinoma (ACC), where it can be mutated or aberrantly active in up to 90% of cases.[2] Preclinical studies have substantiated this mechanistic aspect, demonstrating that CY-101 treatment leads to the downregulation of downstream target genes of the Wnt/β-catenin pathway in relevant tumor models.[10] This secondary mechanism significantly broadens the therapeutic rationale for CY-101, positioning it as a targeted agent for cancers reliant on this signaling cascade. The evolution in the understanding and branding of CY-101 from a primarily lytic peptide to a bifunctional immunotherapy incorporating Wnt/β-catenin inhibition suggests that deeper research has uncovered this significant additional mode of action, enhancing its therapeutic potential.
The dual mechanisms of action attributed to CY-101—direct tumor cell membranolysis and concurrent inhibition of the Wnt/β-catenin signaling pathway—are not merely independent activities but are hypothesized to function synergistically to achieve a more profound and durable anti-tumor effect.[2]
The membranolytic action initiates a cascade of events beneficial for anti-tumor immunity. By rapidly lysing cancer cells, CY-101 facilitates the release of a broad spectrum of tumor-associated antigens (TAAs) and DAMPs directly within the tumor microenvironment.[3] This "antigenic payload" becomes readily available for uptake and processing by antigen-presenting cells (APCs), such as dendritic cells, effectively transforming the tumor into an in situ vaccine. This process is designed to prime and activate a robust, tumor-specific adaptive immune response, including the generation of cytotoxic T lymphocytes (CTLs) capable of recognizing and eliminating remaining cancer cells, potentially including those at distant metastatic sites (abscopal effect).
Concurrently, the inhibition of the Wnt/β-catenin pathway by CY-101 addresses a common mechanism of immune evasion and tumor progression.[2] Aberrant Wnt/β-catenin signaling is often associated with an immunosuppressive TME, characterized by poor T cell infiltration and function. By downregulating this pathway, CY-101 may help to:
The synergy arises because the immune response initiated by membranolysis is likely to be more effective in a TME that has been "reconditioned" by Wnt/β-catenin inhibition. An increased presence of functional CTLs, facilitated by Wnt pathway modulation, can more effectively target and eliminate cancer cells that might have survived the initial lytic insult or those that are newly identified through the antigen presentation process.
This dual action is particularly compelling in the context of combination therapies with immune checkpoint inhibitors (ICIs) like anti-PD-1 antibodies (e.g., pembrolizumab).[3] ICIs work by releasing the "brakes" on existing anti-tumor T cells. CY-101, by increasing antigen presentation and potentially improving T cell infiltration and function through Wnt modulation, could enhance the pool of tumor-reactive T cells available for ICI activation and convert immunologically "cold" tumors (often resistant to ICIs) into "hot," T cell-inflamed tumors that are more susceptible to checkpoint blockade. Preclinical data showing enhanced efficacy of CY-101 when combined with anti-PD-1 therapy support this synergistic hypothesis.[3]
The early pharmacokinetic profile observed in rats, indicating rapid systemic clearance and significant RES uptake [1], strongly influenced the clinical development strategy towards intratumoral administration. This route is ideal for maximizing local drug concentrations, thereby optimizing both the direct lytic effects and the modulation of the local TME, including Wnt pathway activity. This approach also aims to minimize potential systemic toxicities that might be associated with broader exposure to a lytic peptide or a Wnt pathway inhibitor, leveraging the drug's localized action to achieve a systemic immune response.
The preclinical development of CyPep-1/CY-101 has provided a substantial body of evidence supporting its unique mechanisms of action and therapeutic potential. These studies have spanned in vitro characterization of its cytotoxic and mechanistic properties to in vivo assessments of efficacy, pharmacokinetics, and immune modulation in various animal models.
In vitro investigations were crucial in establishing the fundamental cytotoxic activity and selectivity of CyPep-1. The peptide demonstrated a clear dose- and time-dependent toxicity across a panel of human and murine tumor cell lines, including those derived from osteosarcoma, glioma, and mammary carcinoma.[1] For instance, after a 60-minute incubation with 20 µg/mL of CyPep-1, average surviving fractions of cancer cell lines ranged from as low as 7% for the highly sensitive mammary carcinoma line AU565, to 33% for the less sensitive glioblastoma line GaMg.[1] This cytotoxic effect was significantly less pronounced on normal human fibroblasts (HFF1 and HOB cell lines), which showed survival rates of 66% and 44%, respectively, under similar conditions. On average, cancer cell lines exhibited a surviving fraction of 18.6% compared to 56% for normal human cell lines, highlighting a degree of selectivity.[1] This preferential activity against cancer cells is attributed to electrostatic interactions with their negatively charged cell membranes, often resulting from an increased surface exposure of anionic phospholipids like phosphatidylserine.[1]
The membranolytic mechanism was further elucidated through various techniques. Scanning electron microscopy (SEM) visually captured the destructive effects of CyPep-1 on cancer cell membranes, revealing the formation of numerous pores and, with prolonged exposure, complete membrane disintegration.[1] Time-lapse confocal microscopy corroborated these findings, showing extensive loss of cell content into the surrounding medium.[1] Biophysical studies using surface plasmon resonance (SPR) confirmed CyPep-1's binding affinity for liposomes, with a significantly higher affinity observed for negatively charged liposomes compared to neutral ones. Complementary liposome leakage assays demonstrated that CyPep-1 induced a dose-dependent release of encapsulated fluorophore, indicating membrane disruption.[1]
More recent in vitro work focusing on the CY-101 designation and its dual mechanism has shown that it induces dose-dependent cell death in various mouse and human cancer cell lines known to be driven by β-catenin, including colorectal carcinoma (CRC), adrenocortical carcinoma (ACC), and melanoma. This cell death was correlated with the suppression of the Wnt/β-catenin signaling pathway.[10] These findings provide in vitro support for both the direct lytic and Wnt-modulatory actions of the peptide.
The anti-tumor potential of CyPep-1/CY-101 observed in vitro was further investigated in several in vivo animal models:
Early pharmacokinetic (PK) studies of 125I-labeled CyPep-1 were conducted in rats following intravenous (i.v.) administration. These studies revealed a rapid decline in plasma activity, with the most significant drop occurring within the first 5 minutes post-injection, and an estimated plasma half-life of approximately 3.15 minutes.[1] Tissue distribution analysis showed that the peptide was predominantly retained in organs of the reticuloendothelial system (RES), namely the liver and spleen, as well as the kidneys. Conversely, concentrations in other tissues like the lungs, gastrointestinal tract, and muscle were comparatively low. Importantly, extremely low concentrations were detected in central nervous system (CNS) organs, indicating that CyPep-1 did not readily cross the blood-brain barrier.[1]
These PK characteristics—rapid systemic clearance and high RES uptake—led to the initial conclusion that CyPep-1, in its then-current form, was likely not well-suited for systemic administration aiming for widespread tumor targeting.[1] This understanding heavily influenced the subsequent clinical development strategy, which has focused on direct intratumoral injection. This route bypasses the challenges of systemic delivery for such a peptide, maximizes local drug concentration at the tumor site, and is well-aligned with its proposed mechanisms of local tumor lysis and TME modulation.
The use of D-amino acids in the peptide's structure is a key feature contributing to its stability. Unlike peptides made from L-amino acids, D-amino acid peptides are resistant to degradation by common serum proteases.[1] This inherent stability is crucial for maintaining the peptide's integrity and activity in the biological environment, particularly when administered locally where it needs to persist to exert its effects. Preclinical comparisons showed that D-amino acid formulations of the peptide were more potent in their cytotoxic effects than L-amino acid versions [1], underscoring the pharmaceutical advantage of this design choice.
The collective preclinical data, from in vitro selectivity and cytotoxicity to in vivo efficacy and immune modulation, particularly the synergy with anti-PD-1 agents and the Wnt pathway effects, have established a strong scientific foundation for the clinical development of CY-101. The early PK profile also critically shaped the delivery strategy, emphasizing local administration to harness its potent effects while managing potential systemic limitations.
Table 2: Summary of Key Preclinical Studies for CyPep-1 / CY-101
Study Type | Model System | Agent(s) | Key Parameters Investigated | Salient Findings | Reference(s) |
---|---|---|---|---|---|
In Vitro | Various cancer cell lines (osteosarcoma, glioma, mammary carcinoma), normal human fibroblasts | CyPep-1 monotherapy | Cytotoxicity, selectivity, time/dose-dependence, membrane disruption (SEM, confocal), liposome binding/leakage | Selective, dose/time-dependent cytotoxicity to cancer cells; less effect on normal fibroblasts; membrane pore formation and lysis; higher affinity for negatively charged liposomes. | 1 |
In Vitro | Mouse/human β-catenin-driven CRC, ACC, melanoma cell lines | CY-101 monotherapy | Cell death, Wnt/β-catenin pathway suppression | Dose-dependent cell death; suppression of Wnt/β-catenin pathway. | 10 |
In Vivo | 4T1 murine mammary carcinoma allograft model | CyPep-1 monotherapy (intratumoral) | Tumor growth, survival, tumor histology | Significant tumor growth reduction; prolonged survival; extensive tumor necrosis. | 1 |
In Vivo | ACC tumor models (animal) | CY-101 monotherapy | Tumor eradication | Complete eradication of ACC tumors. | 10 |
In Vivo | Various tumor models (general reference) | CyPep-1 + anti-PD-1 | Tumor volume, survival | Synergistic effect: decreased tumor volumes and prolonged survival compared to monotherapies. | 3 |
In Vivo | Immune-refractory CRC and melanoma models | CY-101 + anti-PD-1 | Anti-tumor efficacy, immune landscape modulation (cytokines, CD8+ T cell infiltration), Wnt/β-catenin target gene expression | Significantly enhanced anti-PD-1 efficacy; reshaped tumor immune landscape (increased CD8+ T cell infiltration); downregulation of Wnt/β-catenin downstream target genes; triggered systemic antitumor immune response. | 6 |
Pharmacokinetics | Rats | 125I-CyPep-1 (i.v. injection) | Plasma half-life, tissue distribution, blood-brain barrier penetration | Short plasma half-life (~3.15 min); retention in liver, spleen, kidneys; did not cross blood-brain barrier; suggested unsuitability for systemic administration in its then-current form. | 1 |
Stability | General (peptide design) | CyPep-1 (D-amino acid composition) | Proteolytic stability, potency | D-amino acid composition confers resistance to proteolytic degradation and enhances potency compared to L-amino acid versions. | 1 |
The clinical development of CY-101 is spearheaded by the CICILIA trial (NCT04260529), a multi-faceted study designed to rigorously evaluate its potential in patients with advanced solid cancers. This trial represents a critical step in translating the promising preclinical findings into human therapeutic application.
The CICILIA trial is formally titled "CyPep-1 Injections in Cancer Inducing Lymphocyte Infiltrate Accumulations" or, more descriptively, "A First-in-human, Open-label Dose Escalation Followed by Dose Expansion Phase I/IIa Trial to Evaluate the Safety, Preliminary Efficacy and Pharmacokinetics of Intratumoral CyPep-1 Monotherapy and in Combination With Pembrolizumab in Patients With Advanced Solid Cancers".[3] This Phase I/IIa study employs an open-label design and is structured in distinct phases to systematically assess CY-101.
Objectives: The primary objectives include evaluating the safety and tolerability of intratumorally administered CY-101, determining the maximum tolerated dose (MTD) if applicable, and identifying the Recommended Phase 2 Dose (RP2D) for both monotherapy and combination therapy with pembrolizumab. Secondary objectives encompass assessing preliminary anti-tumor efficacy (effects on injected and distant non-injected lesions), characterizing the pharmacokinetic (PK) and pharmacodynamic (PD) profiles of CY-101, and investigating its local and systemic immunological effects.[3]
Patient Population and Administration: The trial enrolls patients with a variety of advanced solid cancers who have typically undergone extensive prior treatments and have limited therapeutic options.[3] CY-101 is administered directly into malignant tumors via intratumoral injection.[3]
Status, Enrollment, and Locations: The CICILIA trial is ongoing and has progressed through several stages, including dose escalation and various expansion arms. Initial reports mentioned an enrollment target of N=60, with later updates indicating N=75 and a 57-patient cohort for which top-line results were anticipated in the latter half of 2023.[12] Full safety and efficacy data from the completed Phase I/IIa trial are slated for presentation at the European Society for Medical Oncology (ESMO) Congress in 2024.[2] The trial has been conducted at renowned cancer centers across Europe, including sites in The Netherlands (e.g., Netherlands Cancer Institute, Erasmus MC), France, and Spain [4], with plans for expansion into Germany, the UK, and the United States, particularly for the combination studies.[13] The adaptive trial design, incorporating basket cohorts and expansion arms, allows for efficient exploration of CY-101's utility across different tumor types and therapeutic settings based on emerging data.
Part 1 of the CICILIA trial focused on CY-101 monotherapy, involving a dose-escalation phase to establish safety and determine the RP2D. This phase successfully concluded, having reached the planned target dose levels.[12] Early reports indicated that 12 patients were recruited into this initial part, each receiving a minimum of three intratumoral injections of CY-101.[12]
Safety, Tolerability, and RP2D: A consistent and favorable safety profile has been a hallmark of CY-101 monotherapy in these early phases. It has been reported as well-tolerated, with no serious adverse events or dose-limiting toxicities (DLTs) observed during the Part 1 monotherapy dose escalation.[2] This encouraging safety profile allowed for the definition of the RP2D at 20 mg, as reported in data intended for ESMO 2024.[2] The absence of DLTs at effective dose levels is a significant positive, de-risking further development and supporting its use in combination regimens.
Early Efficacy Signals (Monotherapy): Promising early signals of anti-tumor efficacy with CY-101 monotherapy have been consistently reported, aligning with the preclinical proof-of-concept data.[4] Analyses of paired tumor biopsies from treated patients demonstrated induction of cancer cell death in over 70% of cases across various tumor types when administered at the RP2D.[2] This biological activity translated into clinical benefit for several patients with different cancer types.[2] The observation that the most promising responses were seen in patients whose tumors were characterized by aberrant Wnt/β-catenin pathway signaling provides crucial validation for CY-101's dual mechanism and guides patient selection for future studies.[11]
Melanoma Expansion Arm: Building on the encouraging Phase 1 monotherapy data, a Phase 2a expansion arm of the CICILIA trial was initiated to specifically investigate CY-101 monotherapy in patients with advanced melanoma refractory to checkpoint inhibitors. The first patient in this cohort was dosed in February 2023.[4]
A key component of the CICILIA trial involves evaluating CY-101 in combination with pembrolizumab (KEYTRUDA®), leveraging the hypothesis that CY-101's immunogenic cell death and TME modulation can synergize with PD-1 blockade.[3]
Rationale and Collaboration: Preclinical data strongly suggested that CY-101's mechanism of action—inducing tumor cell lysis and releasing tumor antigens—is highly amenable to combination with checkpoint inhibitors.[3] This led to a clinical trial collaboration and supply agreement with MSD.[6]
Study Design and Target Indications: The CICILIA trial was expanded to include a combination arm where CY-101 is administered with pembrolizumab.[3] An initial cohort of 15 patients was planned to primarily evaluate the safety of this combination.[12] Following the safety lead-in, Phase II combination studies are intended to assess efficacy in patients with advanced Head and Neck Squamous Cell Carcinoma (HNSCC), Triple-Negative Breast Cancer (TNBC), and Melanoma.[4] These studies are planned for sites in both the U.S. and Europe.[22]
Status and Timelines: The first patient in the combination expansion phase was dosed in late 2021 or early 2022.[12] Initial results from the safety study of the combination arm were anticipated in early 2022, with the Phase II combination efficacy trials scheduled to commence shortly thereafter.[12]
A formal objective of the CICILIA trial is the assessment of the pharmacokinetic (PK) and pharmacodynamic (PD) properties of CY-101, both as a monotherapy and when administered in combination with pembrolizumab.[3] This includes understanding how the drug is absorbed locally, distributed, and potentially cleared, as well as its biological effects on the tumor and the immune system within treated patients. While these are stated objectives, specific detailed human PK or PD data (e.g., half-life in tumor, systemic exposure, biomarker modulation kinetics) have not been extensively detailed in the currently available public summaries beyond the planned analyses.
Key findings from the CICILIA trial have been and continue to be disseminated through scientific conferences:
The consistent safety profile demonstrated for CY-101 monotherapy in the CICILIA trial is a significant asset. The absence of DLTs in the dose-escalation phase allowed for the determination of an RP2D that exhibits biological activity, instilling confidence for its progression into later-phase studies and, importantly, into combination therapies where managing additive toxicities is crucial. This favorable safety likely reflects some degree of the cancer cell selectivity observed in preclinical models.
Table 3: Overview of the CICILIA Clinical Trial (NCT04260529)
Feature | Description | Source(s) |
---|---|---|
Phase | Phase I/IIa | 3 |
Full Title | "CyPep-1 Injections in Cancer Inducing Lymphocyte Infiltrate Accumulations" / "A First-in-human, Open-label Dose Escalation Followed by Dose Expansion Phase I/IIa Trial to Evaluate the Safety, Preliminary Efficacy and Pharmacokinetics of Intratumoral CyPep-1 Monotherapy and in Combination With Pembrolizumab in Patients With Advanced Solid Cancers" | 3 |
NCT Number | NCT04260529 | 3 |
Sponsor | Cytovation AS | 12 |
Key Objectives | Evaluate safety, tolerability, PK, PD, preliminary efficacy of intratumoral CY-101 (monotherapy & + pembrolizumab), determine RP2D, monitor local/distant tumor effects, assess immunological effects. | 3 |
Study Arms/Cohorts | Monotherapy Dose Escalation; Monotherapy Expansion (e.g., Melanoma refractory to CPIs, Adrenocortical Carcinoma); Combination with Pembrolizumab (Safety lead-in, then HNSCC, TNBC, Melanoma). | 2 |
Interventions | Intratumoral CY-101; Pembrolizumab (KEYTRUDA®) in combination arms. | 3 |
Primary Endpoints | Safety (Adverse Events), Dose-Limiting Toxicities (DLTs), Recommended Phase 2 Dose (RP2D). | 32 |
Key Secondary Endpoints | Objective Response Rate (ORR), Duration of Response (DOR), Pharmacokinetics (PK), Pharmacodynamic (PD) markers (e.g., immune cell infiltration, biomarker changes in biopsies). | 3 (inferred) |
Current Status | Phase I/IIa completed; Full data presentation at ESMO 2024. Expansion arms have been active. | 2 |
Key Locations | The Netherlands, France, Spain, Germany (planned), UK (planned), USA (for combination studies). | 4 |
Table 4: Summary of Clinical Efficacy and Safety Data from the CICILIA Trial (NCT04260529)
Data Source/Presentation | Cohort(s) | Number of Patients (N) (approx.) | CY-101 Dose/RP2D | Key Safety Findings | Key Efficacy Findings | Biomarker Correlations | Source(s) |
---|---|---|---|---|---|---|---|
Cytovation Press Release (Sept 2021) | Monotherapy Dose Escalation (Part 1) | 12 | Target dose reached | Favorable safety, no SAEs or DLTs. | Encouraging early efficacy signals, consistent with preclinical data. | Not detailed. | 12 |
Cytovation Press Release (Oct 2023) / Interim Data Part 2 | Advanced/Metastatic Solid Tumors (Basket trial) | 57 (overall trial progress) | Not specified for this interim release | Excellent safety profile, no DLTs. | Strong early signs of efficacy across solid tumor types. Most promising responses in Wnt/β-catenin aberrant tumors & liver metastases. | Responses correlated with Wnt/β-catenin pathway aberration. | 11 |
Cytovation Press Release / ESMO 2024 Abstract (Sept 2024) | Monotherapy Dose Escalation; Overall Solid Tumors; Adrenocortical Carcinoma (ACC) | Not specified for all cohorts (6 in ACC monotherapy) | RP2D defined at 20 mg | Well tolerated, no DLTs in dose escalation. | >70% cancer cell death in biopsies at RP2D; Clinical benefit in several cancer types. ACC: DCR 50% (3/6), durable responses >6mo (2/6). | ACC responses in patients with β-catenin expression and somatic Wnt/β-catenin pathway mutations. | 2 |
ESMO 2022 (476P Eskens et al.) | Advanced Solid Tumors (Dose Escalation) | Not specified in snippets | Dose escalation ongoing | Not detailed in snippets beyond trial conduct. | Not detailed in snippets. | Not detailed in snippets. | 12 |
A significant strategic direction in the development of CY-101 has emerged with a strong focus on Adrenocortical Carcinoma (ACC) and other malignancies characterized by dysregulation of the Wnt/β-catenin signaling pathway. This focus is underpinned by CY-101's dual mechanism of action, particularly its ability to inhibit this critical oncogenic pathway.
Adrenocortical Carcinoma is a rare and highly aggressive endocrine malignancy with a generally poor prognosis, particularly when diagnosed at an advanced or metastatic stage.[11] Standard treatment options are limited, and there is a significant unmet medical need for novel, effective therapies.[14]
A key molecular characteristic of ACC is the frequent aberrant activation of the Wnt/β-catenin signaling pathway. Estimates suggest that this pathway is dysregulated in a substantial proportion of ACC cases, potentially as high as 90% in some cohorts, often due to mutations in genes like CTNNB1 (encoding β-catenin) or other pathway components.[2] This pathway plays a crucial role in normal adrenal development but its constitutive activation in ACC contributes to tumor cell proliferation, survival, and potentially resistance to conventional therapies. Given that CY-101 has demonstrated an ability to inhibit the Wnt/β-catenin pathway, potentially by activating Axin2, and induce cancer cell death in Wnt-driven preclinical models [2], ACC represents a highly rational target indication. The Wnt/β-catenin pathway is also known to be dysregulated in approximately 20% of all solid tumors, further broadening the potential applicability of CY-101 if this mechanism proves clinically impactful.[11]
Early clinical data from the monotherapy expansion cohorts of the CICILIA trial have provided encouraging evidence for CY-101's activity in ACC patients. As reported in communications leading up to ESMO 2024:
These findings are highly significant as they provide the first clinical validation of CY-101's Wnt/β-catenin inhibitory mechanism in a patient population where this pathway is a known driver. Successfully treating ACC, a notoriously difficult cancer, would offer substantial proof-of-concept for this aspect of CY-101's activity.
Based on the promising early clinical signals in ACC and the strong mechanistic rationale, Cytovation, in collaboration with major cancer research organizations, is advancing CY-101 into a dedicated multinational Phase II clinical trial for patients with ACC.[2]
This trial, which is planned with registrational intent, is a significant step forward. It is expected to commence in late 2025, with initial clinical readouts anticipated in 2026.[6] The collaboration involves Cancer Research UK's Centre for Drug Development (CDD), which will sponsor, design, and deliver the trial across sites in the UK and other European countries, and the Norwegian Cancer Society, which will provide co-funding.[14] Cytovation will be responsible for providing CY-101 for the trial. This collaborative model is a strategic approach to conducting a potentially pivotal trial in a rare disease setting, sharing financial risk and leveraging the extensive expertise and infrastructure of these established organizations. This "fast-to-market" strategy in ACC is intended to provide a clear development path and potentially an earlier approval for CY-101 in an area of high unmet need.[11]
The validation of CY-101's efficacy in ACC, a quintessential Wnt/β-catenin driven cancer, is expected to serve as a "bridgehead" for its expansion into other, more common malignancies where this pathway is also frequently dysregulated.[11] These include, but are not limited to, Colorectal Cancer (CRC), Hepatocellular Carcinoma (HCC), and certain types of liver, uterine, lung, and ovarian cancer.[2]
Preclinical data already support the potential of CY-101 in some of these indications. For instance, studies in immune-refractory models of CRC demonstrated that CY-101 could eliminate tumors and significantly enhance the efficacy of anti-PD-1 therapy, with mechanistic evidence showing downregulation of Wnt/β-catenin target genes.[6] The broader applicability of CY-101 in Wnt-driven cancers is a key aspect of its future development strategy, aiming to bring a novel therapeutic option to a significant number of patients across various tumor types. The correlation of responses with Wnt pathway mutations in ACC patients further suggests that biomarker-based patient selection could be a viable strategy to enrich for responders in future trials across these broader indications.
The regulatory journey and intellectual property (IP) framework are critical components for any novel therapeutic. For CY-101, key milestones have been achieved, particularly with the U.S. Food and Drug Administration (FDA), and a patent portfolio is in place to protect the innovation.
Cytovation has actively engaged with the FDA, leading to important regulatory allowances and designations that facilitate CY-101's clinical development in the United States:
Protecting the intellectual property associated with a novel therapeutic like CY-101 is paramount.
The combination of IND acceptance for broader combination trials and the strategic ODD for ACC demonstrates a thoughtful regulatory approach by Cytovation. The ODD, in particular, acts as a crucial lever, potentially accelerating development for a high-need indication while providing valuable market protection. This, coupled with a foundational patent estate, forms a solid basis for CY-101's continued development and future commercialization.
Table 5: Regulatory Milestones for CyPep-1 / CY-101
Regulatory Action | Regulatory Agency | Date/Year | Indication(s) | Key Implications | Source(s) |
---|---|---|---|---|---|
Investigational New Drug (IND) Application Acceptance | US FDA | May 2022 | Phase II studies of CY-101 in combination with Pembrolizumab (KEYTRUDA®) for Melanoma, Head and Neck Squamous Cell Carcinoma (HNSCC), and Triple-Negative Breast Cancer (TNBC). | Allowed initiation of clinical trials in the U.S. for these combination therapies. | 22 |
Orphan Drug Designation (ODD) | US FDA | Announced by Sept 2024 2 | CY-101 monotherapy for the treatment of Adrenocortical Carcinoma (ACC). | Provides development incentives (e.g., tax credits, fee waivers), increased FDA interaction, and eligibility for seven years of market exclusivity in the U.S. upon approval for ACC. | 2 |
While the primary focus of current development and this report is on CY-101 for oncological indications, the underlying peptide, CyPep-1, was also explored for dermatological applications under a different formulation and designation, CyPep-H1. This earlier exploration provides context to the peptide's broader biological activity and Cytovation's evolving strategic focus.
CyPep-H1 was developed as a topical formulation of CyPep-1 specifically for the treatment of cutaneous warts, which are commonly caused by the human papillomavirus (HPV).[7] The proposed mechanism of action for CyPep-H1 in this context was similar to the lytic action observed for CY-101: the peptide was thought to selectively target HPV-infected (transformed) skin cells, which, like some cancer cells, may present altered membrane characteristics such as increased negative charge.[7] Upon interaction, CyPep-H1 was expected to disrupt the cell membrane of these wart cells, leading to their lysis. This direct cytolytic effect was also hypothesized to trigger a local immune response through the release of viral and cellular antigens, potentially providing long-term protection against wart recurrence.[7]
Cytovation had progressed CyPep-H1 through preclinical development with plans to initiate a Phase I/IIa clinical trial in patients with cutaneous warts, with initial results anticipated in late 2018.[7] Patent filings also reflected this dermatological interest, with claims covering pharmaceutical compositions of CyPep-1 for treating neoplastic lesions, with specific mention of warts.[35]
However, more recent information on Cytovation's pipeline indicates a shift in focus. As of early 2025, the development status for CyPep-1 (or its variants) for skin disorders and warts was listed as "No development reported" by AdisInsight.[8] This suggests a deprioritization of the dermatological program in favor of the more advanced and perhaps more strategically compelling oncology indications being pursued with CY-101. The significant investment, high-profile collaborations (MSD, Cancer Research UK), and regulatory milestones (IND acceptance for combination oncology trials, ODD for ACC) achieved for CY-101 in cancer underscore this strategic pivot. While the early work on CyPep-H1 demonstrated the peptide's lytic capabilities and potential for topical application, the company's resources and efforts appear now to be concentrated on harnessing its dual mechanism of action (membranolysis and Wnt/β-catenin inhibition) for systemic and locally advanced cancers via intratumoral administration. This strategic shift is common in pharmaceutical development, where companies prioritize assets and indications based on emerging data, market potential, and unmet medical need.
CY-101 (CyPep-1) has emerged as a distinctive investigational agent in the oncology landscape, characterized by its novel peptide nature and, more importantly, its dual mechanism of action. Its journey from a concept of a cationic lytic peptide to a clinical-stage bifunctional immunotherapy targeting both cancer cell membranes and the Wnt/β-catenin pathway reflects a data-driven evolution in understanding its therapeutic potential.
The primary therapeutic potential of CY-101 lies in its first-in-class status as a bifunctional immunotherapy.[2] This is not merely a peptide that lyses cells; its ability to concurrently inhibit the oncogenic Wnt/β-catenin pathway sets it apart.[2] This dual attack—direct tumor destruction leading to in situ vaccination and modulation of a key cancer signaling pathway implicated in growth and immune evasion—offers a multi-pronged strategy against complex diseases like cancer.
Key differentiating features include:
The clinical validation of CY-101's efficacy in ACC, a cancer type with very high Wnt/β-catenin pathway dysregulation, serves as a strong proof-of-concept for its Wnt-inhibitory mechanism.[2] This opens the door for investigating CY-101 in a broader range of solid tumors where this pathway is also a known oncogenic driver. Approximately 20% of all solid tumors exhibit aberrant Wnt/β-catenin signaling [11], including highly prevalent cancers such as:
The ACC program, particularly the Phase II trial with registrational intent, could act as a strategic "bridgehead," providing robust clinical data and potentially an earlier market entry that could then be leveraged to expand into these larger, Wnt-driven cancer indications.[11]
Challenges:
Opportunities:
Future research for CY-101 will likely focus on several key areas:
The development path of CY-101, particularly its focus on the Wnt/β-catenin pathway, is ambitious given the historical challenges in drugging this pathway due to its role in normal tissue homeostasis.[24] However, the localized delivery strategy and the encouraging early safety data from the CICILIA trial [2] suggest that Cytovation may have found a viable approach to harness its potential while mitigating risks. Its success will ultimately depend on robust data from ongoing and planned later-phase trials, particularly in demonstrating clear differentiation and benefit in a competitive immuno-oncology landscape.
CyPep-1, clinically developed as CY-101 by Cytovation AS, represents a compelling and innovative approach in the field of oncology. Its journey from an academic peptide concept to a clinical-stage asset undergoing evaluation in advanced solid tumors underscores a responsive, data-driven development strategy. The peptide's unique D-amino acid structure confers critical stability, while its dual mechanism of action—direct, selective tumor cell membranolysis leading to immunogenic cell death, and concurrent inhibition of the oncogenic Wnt/β-catenin signaling pathway—positions it as a first-in-class bifunctional immunotherapy.
Preclinical studies have consistently demonstrated CY-101's potent anti-tumor activity, both as a monotherapy and in synergistic combination with checkpoint inhibitors, particularly in models of Wnt-driven cancers. These studies have also provided a strong mechanistic rationale for its effects on the tumor microenvironment, including enhanced T-cell infiltration and modulation of key oncogenic signaling.
The Phase I/IIa CICILIA trial has been instrumental in translating these preclinical findings into the human setting. Early clinical data have been encouraging, highlighting a favorable safety profile for CY-101 with no dose-limiting toxicities observed in the monotherapy dose-escalation phase, and the establishment of a recommended Phase 2 dose. More significantly, promising signals of clinical efficacy have emerged, particularly in patients with Adrenocortical Carcinoma whose tumors harbor Wnt/β-catenin pathway dysregulation. This has led to a strategic prioritization of ACC, supported by an Orphan Drug Designation from the US FDA and significant international collaborations for a dedicated Phase II trial with registrational intent.
The strategic adaptation from a broadly lytic peptide to a more defined dual-mechanism immunotherapy targeting specific cancer vulnerabilities, such as Wnt pathway dependence, reflects a maturing understanding of CY-101's potential. While challenges related to intratumoral delivery in widespread disease and the long-term management of complex biological responses remain, the opportunities are substantial. CY-101 offers a novel therapeutic modality for cancers with high unmet medical need, particularly those driven by Wnt/β-catenin signaling and those that may be refractory to existing immunotherapies.
In conclusion, CY-101 stands out as a promising investigational agent with a unique profile. Its continued clinical development, especially the upcoming data from the completed CICILIA trial and the focused ACC Phase II study, will be critical in further defining its role and potential impact on the future landscape of cancer treatment. If successful, CY-101 could offer a significant new therapeutic option for patients with difficult-to-treat solid tumors.
Published at: May 23, 2025
This report is continuously updated as new research emerges.