Small Molecule
C28H26N4O4
112953-11-4
7-Hydroxystaurosporine, known predominantly by its code name UCN-01, is an investigational small molecule antineoplastic agent belonging to the indolocarbazole class of alkaloids.[1] Initially isolated from
Streptomyces sp. and subsequently developed as a synthetic derivative of the natural product staurosporine, UCN-01 was first characterized as a selective inhibitor of Protein Kinase C (PKC).[1] Further investigation, however, revealed it to be a potent, non-selective, ATP-competitive inhibitor of a broad spectrum of protein kinases critical to cell cycle control and survival signaling pathways.[5] Its primary molecular targets include multiple isoforms of PKC, various Cyclin-Dependent Kinases (CDKs), the pivotal DNA damage checkpoint kinase Chk1, and key components of the PI3K/Akt survival pathway, such as PDK1 and Akt itself.[7]
The compound's cellular pharmacology is characterized by a notable dual mechanism of action. As a monotherapy, it induces a cytostatic G1/S phase cell cycle arrest by inhibiting G1 CDKs and preventing the phosphorylation of the Retinoblastoma protein.[1] In contrast, when administered following DNA-damaging agents like cisplatin or radiation, it acts as a potent cytotoxic sensitizer by abrogating the S and G2 checkpoints, primarily through Chk1 inhibition. This forces damaged cells into a lethal mitosis, providing a strong rationale for its development in combination therapies.[8]
Despite this compelling preclinical profile, the clinical development of UCN-01 was profoundly complicated by a unique and challenging pharmacokinetic profile. The drug exhibits exceptionally high-affinity, saturable binding to the plasma protein α1-acid glycoprotein (AAG), which acts as a circulating reservoir.[8] This interaction results in an unusually prolonged terminal half-life of over 400 hours, nonlinear clearance, and a significant disconnect between the measurable total drug concentration and the pharmacologically active unbound fraction, which constitutes only about 1% of the total.[13] This "AAG trap" created substantial interpatient variability and made rational dosing and the establishment of a clear dose-response relationship exceptionally difficult.
UCN-01 was evaluated in numerous Phase I and II clinical trials across a wide array of hematologic malignancies and solid tumors, both as a single agent and in combination with standard chemotherapeutics.[15] However, its potent preclinical activity failed to translate into significant clinical efficacy, with objective responses being rare and many trials being terminated without progressing to Phase III.[18] The safety profile was marked by dose-limiting toxicities, most notably hyperglycemia and hypotension.[8] The hyperglycemia was mechanistically linked to the drug's intended on-target inhibition of the PDK1/Akt pathway, a critical component of insulin signaling, thereby representing a classic case of on-target, off-tumor toxicity that created an inherently narrow therapeutic window.[22]
Ultimately, 7-Hydroxystaurosporine serves as a pivotal case study in modern oncology drug development. Its journey from a promising preclinical candidate to a clinical failure illustrates the formidable challenges of translating a potent but non-selective kinase inhibitor into a viable therapeutic. The convergence of complex pharmacokinetics, a narrow therapeutic index defined by mechanism-based toxicity, and a lack of compelling clinical efficacy created an insurmountable barrier to its successful development.
7-Hydroxystaurosporine is a complex heterocyclic small molecule that has been extensively characterized. Its identity is established through a variety of nomenclature systems, registry numbers, and detailed structural and physical properties.
The compound is most widely recognized in scientific and clinical literature by the code name UCN-01.[24] It has also been assigned other developmental codes, including KRX-0601 and KW2401, as well as the National Service Center number NSC 638850.[5]
Due to its intricate, polycyclic, and stereochemically rich structure, its formal IUPAC (International Union of Pure and Applied Chemistry) name is exceptionally complex and has been represented in multiple forms. One such systematic name is (5R,7R,8R,9S,14S)-14-hydroxy-8-methoxy-9-methyl-7-(methylamino)-6,7,8,9,14,15-hexahydro-5H,16H-17-oxa-4b,9a,15-triaza-5,9-methanodibenzo[b,h]cyclonona[jkl]cyclopenta[e]-as-indacen-16-one.[5] Another is
(3R,9S,10R,11R,13R)-2,3,10,11,12,13-hexahydro-3-hydroxy-10-methoxy-9-methyl-11-(methylamino)-9,13-epoxy-1H,9H-diindolo[1,2,3-gh:3',2',1'-lm]pyrrolo[3,4-j]benzodiazonin-1-one.[7]
Key registry numbers that provide unambiguous identification include its CAS (Chemical Abstracts Service) Number, 112953-11-4, and its DrugBank accession ID, DB01933.[5] In public chemical databases, it is cataloged under PubChem Compound ID (CID) 72271.[27]
The molecular formula for 7-Hydroxystaurosporine is consistently reported as C28H26N4O4.[5] This corresponds to a molecular weight of approximately 482.53 Da and an exact mass of 482.1954 Da.[3] Its elemental composition is approximately 69.70% carbon, 5.43% hydrogen, 11.61% nitrogen, and 13.26% oxygen.[5]
The complex three-dimensional structure of the molecule is captured by unique structural identifiers. The InChIKey, a hashed version of the standard InChI string, is reported as PBCZSGKMGDDXIJ-HQCWYSJUSA-N, which encodes its specific stereochemistry.[7] Various SMILES (Simplified Molecular Input Line Entry Specification) strings are also available to represent its connectivity and stereochemistry in a linear format.[2]
7-Hydroxystaurosporine is classified as an indolocarbazole alkaloid, a class of compounds known for their biological activity, particularly as protein kinase inhibitors.[1] Although it was originally isolated from the culture broth of a bacterial strain,
Streptomyces sp., it is most accurately described as a semi-synthetic or synthetic derivative of the parent natural product, staurosporine.[4] This distinction is crucial, as UCN-01 represents a deliberate chemical modification of a natural scaffold to improve its pharmacological properties. Structurally, it is closely related to other staurosporine-derived kinase inhibitors and is listed as a potential impurity in the manufacturing of Midostaurin.[31]
As a solid, 7-Hydroxystaurosporine appears as a light yellow powder.[3] Its solubility characteristics are typical for a lipophilic organic molecule; it is soluble in organic solvents such as dimethyl sulfoxide (DMSO) at concentrations greater than 5 mg/mL and in ethanol at 1 mg/mL, but it is considered insoluble in water.[3] For research and storage, the compound is stable if kept dry, dark, and refrigerated (0-4 °C for short-term) or frozen (-20 °C for long-term), with a shelf life exceeding five years under proper conditions.[3] Analysis of its structure against Lipinski's Rule of Five, a common measure of "drug-likeness," shows zero or one violation, suggesting that it possesses physicochemical properties generally favorable for membrane permeability and potential oral bioavailability, although it was ultimately developed as an intravenous agent.[9]
A summary of its key chemical and physical properties is provided in Table 1.
Table 1. Chemical and Physical Properties of 7-Hydroxystaurosporine (UCN-01)
Property | Value | Source(s) |
---|---|---|
Common Name | 7-Hydroxystaurosporine | 1 |
Synonyms/Codes | UCN-01, KRX-0601, KW2401, NSC 638850 | 5 |
DrugBank ID | DB01933 | 15 |
CAS Number | 112953-11-4 | 5 |
PubChem CID | 72271 | 27 |
Molecular Formula | C28H26N4O4 | 5 |
Molecular Weight | 482.53 Da | 3 |
IUPAC Name | (5R,7R,8R,9S,14S)-14-hydroxy-8-methoxy-9-methyl-7-(methylamino)-6,7,8,9,14,15-hexahydro-5H,16H-17-oxa-4b,9a,15-triaza-5,9-methanodibenzo[b,h]cyclonona[jkl]cyclopenta[e]-as-indacen-16-one | 5 |
InChIKey | PBCZSGKMGDDXIJ-HQCWYSJUSA-N | 7 |
Appearance | Light yellow powder | 3 |
Solubility | DMSO: >5 mg/mL; Ethanol: 1 mg/mL; Water: Insoluble | 3 |
Storage Conditions | Protect from light; Store at -20 °C (long-term) | 3 |
The antineoplastic activity of 7-Hydroxystaurosporine (UCN-01) stems from its ability to modulate fundamental cellular processes, including signal transduction, cell cycle progression, and apoptosis. Its mechanism is multifaceted, defined by its broad inhibition of multiple protein kinase families, which in turn leads to profound and context-dependent effects on cancer cells.
UCN-01 is a cell-permeable agent that functions as a reversible, ATP-competitive inhibitor of a wide array of protein kinases.[6] Its indolocarbazole scaffold is structurally analogous to the adenine moiety of ATP, allowing it to bind to the highly conserved ATP-binding pocket within the catalytic domain of many kinases, thereby blocking their phosphotransferase activity.[34] Although initially investigated for its selectivity toward Protein Kinase C (PKC), subsequent profiling revealed it to be a non-specific inhibitor with potent activity against several key kinase families that are frequently dysregulated in cancer.[1]
The primary kinase families targeted by UCN-01 include:
The spectrum of kinases inhibited by UCN-01 and their corresponding inhibitory concentrations are summarized in Table 2.
Table 2. Target Kinases and Associated IC50 Values for UCN-01
Target Kinase | Kinase Family | IC50 (nM) | Source(s) |
---|---|---|---|
PDK1 | Survival Kinase | 6 | 7 |
Chk1 | Checkpoint Kinase | 7 | 6 |
PKCα | Protein Kinase C | 29 | 6 |
PKCγ | Protein Kinase C | 30 | 6 |
Cdk4 | Cyclin-Dependent Kinase | 32 | 1 |
PKCβ | Protein Kinase C | 34 | 6 |
Cdk1 | Cyclin-Dependent Kinase | 50 | 6 |
Cdk6 | Cyclin-Dependent Kinase | 58 | 1 |
Cdk2 | Cyclin-Dependent Kinase | 300-600 | 1 |
PKCϵ | Protein Kinase C | 530 | 6 |
PKCδ | Protein Kinase C | 590 | 6 |
Chk2 | Checkpoint Kinase | 1040 | 6 |
The pleiotropic kinase inhibition profile of UCN-01 results in a complex and context-dependent impact on cell cycle regulation. This dual functionality provided the basis for its investigation as both a monotherapy and a combination agent.
When administered as a single agent, UCN-01 primarily functions as a cytostatic drug, inducing a robust cell cycle arrest at the G1/S transition.[1] This effect is a direct consequence of its inhibition of G1-phase CDKs, particularly Cdk2, Cdk4, and Cdk6.[1] These kinases are responsible for phosphorylating and inactivating the Retinoblastoma tumor suppressor protein (pRb). By inhibiting these CDKs, UCN-01 prevents pRb hyperphosphorylation, keeping it in its active, hypophosphorylated state. Active pRb binds to and sequesters E2F transcription factors, thereby blocking the expression of genes essential for DNA replication and entry into S phase.[1] This G1 arrest is also associated with the induction of CDK inhibitor proteins such as p21.[8]
In stark contrast, when UCN-01 is used in combination with DNA-damaging agents like cisplatin or ionizing radiation, its primary role shifts from inducing arrest to abrogating it. DNA damage normally activates cellular checkpoints in the S and G2 phases to halt the cell cycle and allow time for repair.[8] UCN-01 potently overrides these checkpoints.[4] This is achieved through its powerful inhibition of Chk1, the central kinase in the G2 checkpoint pathway.[7] Following DNA damage, Chk1 would normally phosphorylate and inactivate the Cdc25 family of phosphatases. By inhibiting Chk1, UCN-01 ensures that Cdc25 remains active. Active Cdc25 then removes inhibitory phosphates from Cdk1, leading to its premature activation and forcing the cell to enter mitosis despite the presence of extensive DNA damage. This process, often termed "mitotic catastrophe," is a lethal event and is the basis for the synergistic cytotoxicity observed when UCN-01 is combined with genotoxic therapies.[8] This mechanism explains the critical sequence-dependency observed in preclinical and clinical studies, where the DNA-damaging agent must be administered
before UCN-01 to establish the checkpoint that UCN-01 can then abrogate.[19]
In addition to its effects on the cell cycle, UCN-01 is a potent inducer of apoptosis, or programmed cell death, in a wide range of cancer cell lines.[5] This apoptotic response is mediated through the activation of caspases and other serine proteases.[8] Importantly, the induction of apoptosis by UCN-01 appears to be independent of the functional status of the p53 tumor suppressor protein.[8] This is a highly significant therapeutic feature, as p53 is mutated and inactivated in over half of all human cancers, often conferring resistance to conventional chemotherapy. The ability of UCN-01 to bypass the need for functional p53 to induce cell death made it an attractive candidate for treating refractory, p53-deficient tumors.
The clinical pharmacology of UCN-01 is highly unusual and is dominated by a single, overriding factor: its extensive and high-affinity binding to the plasma protein α1-acid glycoprotein (AAG). This interaction dictates the drug's distribution, clearance, and half-life, and created profound challenges for its clinical development.
UCN-01 binds with extremely high affinity (association constant, Ka, of 799×106 L/mol) to AAG.[13] AAG is an acute-phase reactant protein, meaning its plasma concentrations are not constant but can increase significantly in patients with cancer or inflammatory conditions.[13] This binding has several critical pharmacokinetic consequences:
This combination of factors created what can be described as a pharmacokinetic "trap." To achieve a therapeutically effective concentration of unbound drug at the tumor site, a very high total drug dose must be administered. However, this high total dose also increases the unbound concentration systemically, elevating the risk of on-target toxicities in normal tissues and creating a very narrow, if not nonexistent, therapeutic window.
The clinical development program for UCN-01 was extensive, reflecting its strong preclinical rationale. It was investigated across a wide range of tumor types, both as a single agent and in combination with numerous cytotoxic therapies. However, this broad investigation did not culminate in a successful therapy, with development largely ceasing after Phase II trials.
The impetus for clinical trials was strong. UCN-01 demonstrated potent antiproliferative activity against a diverse panel of human cancer cell lines in vitro, with IC50 values often in the nanomolar range.[43] This activity translated to in vivo efficacy in multiple human tumor xenograft models, including breast, lung, and head and neck cancers.[8] A key factor driving its development was its unique pattern of activity in the National Cancer Institute's 60-cell line screen, which was distinct from all other known classes of anticancer drugs, suggesting a novel mechanism of action.[8] Furthermore, its ability to potently abrogate DNA damage checkpoints provided a compelling rationale for its use as a sensitizer for conventional chemotherapy and radiation.[6]
UCN-01 was evaluated in numerous Phase I and Phase II clinical trials sponsored by the National Cancer Institute (NCI) and other groups. The investigations spanned both hematologic malignancies and solid tumors.
Despite the breadth of this clinical program, many of the trials were either terminated early or were completed Phase I dose-finding studies that did not lead to further development.[16] No Phase III trials were initiated, indicating that the drug did not show sufficient promise to warrant large-scale, pivotal studies.[18] A summary of key clinical trials is presented in Table 3.
Table 3. Summary of Key Clinical Trials for 7-Hydroxystaurosporine (UCN-01)
NCT Identifier | Indication(s) | Phase | Status | Combination Agent(s) | Purpose/Brief Description |
---|---|---|---|---|---|
NCT00045513 | Chronic Lymphocytic Leukemia, Lymphoma | 1/2 | Completed | Fludarabine | To evaluate the combination in patients with CLL or lymphocytic lymphoma.15 |
NCT00082017 | T-Cell Lymphomas | 2 | Terminated | None (Monotherapy) | To evaluate UCN-01 in patients with relapsed or refractory T-cell lymphomas.32 |
NCT00012194 | Advanced Malignant Solid Tumors | 1 | Terminated | Cisplatin | Dose-escalation study to determine the MTD of cisplatin combined with UCN-01.16 |
NCT00301938 | Acute Leukemia, CML, MDS | 1 | Active, not recruiting | Perifosine | To determine the MTD of UCN-01 when given with perifosine in hematologic malignancies.45 |
NCT00042861 | Metastatic/Unresectable Solid Tumors | 1 | Completed | Fluorouracil, Leucovorin | To evaluate the combination in patients with advanced solid tumors.17 |
NCT00045747 | Metastatic Pancreatic Cancer | 2 | Completed | Fluorouracil | To evaluate the combination in patients with metastatic pancreatic cancer.47 |
NCT00036777 | Advanced Solid Tumors | 1 | Completed | Carboplatin | Dose-escalation study to determine the MTD of the combination.17 |
NCT00045500 | Solid Tumors, Lymphoma | 1 | Completed | Prednisone | To assess the safety and MTD of UCN-01 combined with prednisone.21 |
The overall clinical efficacy of UCN-01 was disappointing and did not reflect its potent preclinical activity.
Ultimately, the lack of a clear and consistent efficacy signal, coupled with the significant pharmacokinetic and safety challenges, led to the discontinuation of its clinical development.
The clinical use of UCN-01 was associated with a distinct and challenging toxicity profile. The adverse events were often mechanism-based and contributed significantly to the drug's narrow therapeutic window.
Across numerous Phase I trials, a consistent pattern of dose-limiting toxicities (DLTs) emerged:
Common, non-dose-limiting adverse events included Grade 1-2 nausea, vomiting, headache, fatigue, and anorexia, which were generally manageable.[8] When used as a single agent, UCN-01 had minimal hematologic toxicity. However, when combined with myelosuppressive chemotherapies, expected hematologic toxicities such as neutropenia and thrombocytopenia were observed.[14]
Of particular note, a fatal case of Stevens-Johnson syndrome, a severe and rare cutaneous reaction, was reported in a patient receiving UCN-01 in combination with fludarabine, underscoring the potential for severe idiosyncratic toxicities.[46]
The most prominent toxicity, hyperglycemia, is not an off-target side effect but rather a direct consequence of UCN-01's intended mechanism of action. The molecular steps underlying this adverse event have been well-elucidated:
This phenomenon represents a textbook example of on-target, off-tumor toxicity. The very mechanism that makes UCN-01 a potential anticancer agent (inhibition of the Akt survival pathway) is the same mechanism that causes its dose-limiting toxicity in normal tissues (disruption of glucose homeostasis). This creates an inherent and formidable challenge in separating therapeutic effects from adverse effects, leading to a very narrow therapeutic index.
To fully appreciate the rationale for the development of UCN-01, it is essential to compare it to its parent compound, staurosporine. While structurally similar, key pharmacological differences made UCN-01 a more viable candidate for clinical investigation.
Staurosporine is a natural product renowned in cell biology research as a highly potent but notoriously non-selective or "promiscuous" protein kinase inhibitor.[34] It inhibits hundreds of kinases with high affinity, making it an excellent tool for inducing apoptosis in vitro but far too toxic for systemic use as a therapeutic agent.
UCN-01 is a hydroxylated derivative of staurosporine.[54] The addition of the 7-hydroxyl group creates new opportunities for hydrogen bonding within the kinase active site, which alters its kinase inhibition profile.[34] While still a broad-spectrum inhibitor, UCN-01 exhibits a different pattern of selectivity. For example, it shows a clear preference for Ca²⁺-dependent PKC isoforms over Ca²⁺-independent ones, a distinction that is less pronounced with staurosporine.[36]
However, the most critical distinction between the two compounds lies in their therapeutic window. Both staurosporine and UCN-01 are capable of abrogating DNA damage-induced S and G2 checkpoints.[12] The crucial difference is the concentration at which this effect occurs relative to their inherent cytotoxicity. Staurosporine abrogates these checkpoints only at concentrations that are, by themselves, toxic to the cells.[12] In contrast, UCN-01 was uniquely able to achieve checkpoint abrogation and sensitize cancer cells to cisplatin at concentrations that were non-cytotoxic when used alone.[12]
This ability to function as a chemosensitizer within a non-toxic concentration range was the single most important pharmacological advantage of UCN-01 over staurosporine. It provided the fundamental rationale for selecting UCN-01 for clinical development, particularly in combination therapies, as it represented a refined version of staurosporine's power with a potentially manageable safety profile.
7-Hydroxystaurosporine (UCN-01) emerged from a rational drug discovery effort to harness the potent kinase-inhibiting properties of the natural product staurosporine while improving its therapeutic index. Its compelling and elegant dual mechanism of action—inducing cytostatic G1 arrest as a single agent and promoting cytotoxic mitotic catastrophe in combination with genotoxic agents—provided a robust preclinical foundation for its development as a novel anticancer agent.
However, the transition from preclinical promise to clinical success was ultimately thwarted by a convergence of three formidable and interconnected hurdles that are emblematic of the challenges in modern oncology drug development:
In conclusion, 7-Hydroxystaurosporine stands as an important and instructive case study. It underscores the critical principle that potent in vitro activity against a validated cancer target is, by itself, insufficient for clinical success. A viable therapeutic agent must also possess a predictable pharmacokinetic profile, a manageable safety profile, and a therapeutic window wide enough to allow for effective target modulation in the tumor without causing unacceptable harm to the patient. UCN-01, despite its potent and scientifically interesting mechanism, ultimately failed to meet these essential clinical criteria, halting its journey to becoming an approved anticancer drug.
Published at: September 16, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.