Small Molecule
C27H33N3O5
747412-64-2
Luminespib (AUY922) is an investigational, third-generation, fully synthetic small molecule inhibitor of Heat Shock Protein 90 (Hsp90). Identified chemically as a resorcinol-containing isoxazole amide, Luminespib was developed to overcome the pharmaceutical and toxicological limitations of earlier ansamycin-based Hsp90 inhibitors. By competitively binding to the N-terminal ATP pocket of Hsp90 with high affinity, it potently disrupts the chaperone's function, leading to the proteasomal degradation of a wide array of oncogenic client proteins. Preclinical studies demonstrated a powerful and broad-spectrum antineoplastic profile, characterized by the inhibition of critical cancer signaling pathways, induction of apoptosis, and anti-angiogenic effects across numerous cancer cell lines and xenograft models.
This promising preclinical profile propelled Luminespib into a comprehensive clinical development program, spearheaded by Novartis, investigating its efficacy in various advanced solid malignancies. Clinical trials revealed a nuanced and context-dependent activity profile. The most significant clinical benefit was observed in patient populations with tumors driven by strong "oncogene addiction" to highly Hsp90-dependent proteins, most notably in anaplastic lymphoma kinase (ALK)-rearranged non-small cell lung cancer (NSCLC), where a meaningful objective response rate was achieved. Modest activity was also seen in other molecularly defined subsets, including EGFR-mutant NSCLC and HER2-positive breast cancer. However, the broad-spectrum activity anticipated from preclinical data did not materialize in the clinic, with minimal to no efficacy observed in other settings, such as KRAS-mutant NSCLC.
The clinical utility of Luminespib was further constrained by a challenging on-target toxicity profile. The most common treatment-related adverse events included diarrhea, fatigue, and nausea. Critically, a high incidence of reversible but dose-limiting ocular toxicities, such as night blindness and blurred vision, was consistently reported. This safety profile narrowed the therapeutic window, illustrating a fundamental paradox wherein the drug's high potency was directly linked to its dose-limiting toxicities in sensitive normal tissues.
In December 2014, Novartis ceased all development of Luminespib, citing the failure to meet primary efficacy endpoints in a key Phase II trial in NSCLC. The decision was likely multifactorial, reflecting the drug's niche efficacy, its challenging safety profile, and an evolving competitive landscape with more effective and better-tolerated targeted agents. Although Luminespib did not achieve regulatory approval, its development provided invaluable insights into the clinical translation of Hsp90 inhibition, confirming the validity of the target in humans while highlighting the critical challenges of oncogene dependency and on-target toxicity that continue to shape the future of this therapeutic class.
Heat Shock Protein 90 (Hsp90) is a highly conserved and ubiquitously expressed molecular chaperone that is essential for maintaining cellular proteostasis.[1] It functions as a key component of a multi-protein chaperone complex that facilitates the proper folding, conformational maturation, stability, and function of a large and diverse group of substrate proteins, referred to as the Hsp90 "clientele".[3] In normal cells, Hsp90 constitutes 1-2% of total cytosolic protein and plays a critical housekeeping role in signal transduction, cell cycle regulation, and cellular responses to stress.[5]
The role of Hsp90 is fundamentally subverted in malignant cells. Cancer is characterized by the accumulation of mutated, overexpressed, or constitutively active oncoproteins that drive the hallmark capabilities of malignancy, such as sustained proliferative signaling, evasion of apoptosis, limitless replicative potential, and angiogenesis.[2] Many of these key oncoproteins—including receptor tyrosine kinases (e.g., EGFR, HER2, ALK, MET), signaling kinases (e.g., RAF, AKT), and transcription factors (e.g., mutant p53, HIF-1α)—are obligate clients of the Hsp90 chaperone machinery.[2] Cancer cells exhibit a unique dependence, or "addiction," to Hsp90 to buffer the proteotoxic stress caused by this aberrant proteome and to maintain the functional integrity of the oncoproteins upon which their survival depends.[8] Consequently, Hsp90 is frequently overexpressed (2- to 10-fold) in tumor cells compared to normal tissues and exists in a high-affinity, activated multi-chaperone complex state.[5] This differential state provides a therapeutic window, making Hsp90 an attractive and rational target for cancer therapy, as its inhibition can theoretically lead to the simultaneous degradation of multiple oncoproteins, thereby collapsing several oncogenic signaling pathways at once.[7]
The therapeutic potential of Hsp90 inhibition was first discovered with the natural product ansamycin antibiotics, such as geldanamycin and its semi-synthetic derivative 17-allylamino-17-demethoxygeldanamycin (17-AAG, tanespimycin).[12] These first-generation inhibitors validated Hsp90 as a druggable target by demonstrating that binding to the N-terminal ATP pocket of Hsp90 leads to client protein degradation and antitumor activity.[12] However, their clinical development was significantly hampered by major liabilities, including poor aqueous solubility, challenging formulations, significant hepatotoxicity associated with their benzoquinone moiety, and a metabolic dependence on the enzyme NAD(P)H:quinone oxidoreductase 1 (NQO1) for activation, leading to variable patient responses.[12]
These limitations spurred the development of second- and third-generation, fully synthetic Hsp90 inhibitors designed to overcome these issues. Luminespib (AUY922) emerged as a leading candidate from this effort. As a member of the resorcinol-containing isoxazole amide class, Luminespib was rationally designed to offer significant advantages over its predecessors. These include high aqueous solubility, independence from NQO1 metabolism, a lack of the quinone-related hepatotoxicity, and substantially greater potency, with binding affinities and cellular activities in the low nanomolar range.[12] This improved pharmacological profile positioned Luminespib as a promising agent to fully evaluate the clinical hypothesis of Hsp90 inhibition in cancer.
The precise identification and characterization of an investigational compound are foundational for its scientific and clinical evaluation. Luminespib has been described under multiple names and associated with several registry numbers, primarily distinguishing between its free base form and its various salts used in research and formulation.
The compound is most widely known by its developmental code, AUY922 (or NVP-AUY922, reflecting its development by Novartis), and its International Nonproprietary Name (INN), Luminespib.[18] Another code, VER-52296, originates from its discovery by Vernalis.[17]
A critical point of clarification relates to its Chemical Abstracts Service (CAS) numbers. The CAS number for the active moiety, or free base, is 747412-49-3.[18] The CAS number provided in the user query, 747412-64-2, corresponds specifically to the hydrochloride (HCl) salt of the compound.[17] Other salt forms, such as the mesylate, have also been synthesized and are associated with distinct CAS numbers.[17] For regulatory and database tracking, it is also assigned DrugBank ID DB11881.[24]
Luminespib is a derivative of 4,5-diarylisoxazole.[17] The formal IUPAC name for the free base (CAS 747412-49-3) is 5--N-ethyl-4-{4-[(morpholin-4-yl)methyl]phenyl}-1,2-oxazole-3-carboxamide.[18]
The molecular formula of the free base is C26H31N3O5, with a corresponding molecular weight of approximately 465.55 g/mol.[21] The hydrochloride salt (CAS 747412-64-2) has a slightly different reported structure in some sources, featuring a tert-butyl group instead of an isopropyl group, leading to a molecular formula of
C27H34ClN3O5 and a molecular weight of 516.04 g/mol.[17] This discrepancy likely reflects different synthetic variants, though the isopropyl version is the most commonly cited structure in pharmacological literature.
Luminespib is typically supplied as a white to light yellow solid powder.[18] Its solubility profile is a key feature that distinguishes it from first-generation inhibitors. It is soluble in organic solvents such as dimethyl sulfoxide (DMSO) and ethanol but is poorly soluble in aqueous solutions.[19] For clinical administration, an optimized salt was formulated for intravenous infusion.[30] Recommended storage conditions for the solid compound are dry, dark, and refrigerated at 0-4 °C for short-term or frozen at -20 °C for long-term stability.[17]
Table 1: Summary of Chemical and Physical Properties of Luminespib (AUY922) | ||
---|---|---|
Property | Value | Form / Source |
Generic Name (INN) | Luminespib | 21 |
Developmental Codes | AUY922, NVP-AUY922, VER-52296 | 17 |
DrugBank ID | DB11881 | 24 |
CAS Number | 747412-49-3 | Free Base 18 |
747412-64-2 | Hydrochloride (HCl) Salt 17 | |
IUPAC Name | 5--N-ethyl-4-{4-[(morpholin-4-yl)methyl]phenyl}-1,2-oxazole-3-carboxamide | Free Base 21 |
Molecular Formula | C26H31N3O5 | Free Base 21 |
Molecular Weight | 465.55 g/mol | Free Base 21 |
SMILES Code | CCNC(=O)C1=NOC(=C1C2=CC=C(C=C2)CN3CCOCC3)C4=CC(=C(C=C4O)O)C(C)C | Free Base 25 |
InChIKey | NDAZATDQFDPQBD-UHFFFAOYSA-N | Free Base 21 |
Appearance | White to light yellow solid powder | 18 |
Solubility | Soluble in DMSO, Ethanol; Insoluble in H₂O | 19 |
The extensive preclinical characterization of Luminespib established its mechanism as a highly potent and selective inhibitor of Hsp90, elucidating the downstream molecular events that confer its broad antitumor activity.
Luminespib exerts its pharmacological effect by directly targeting the N-terminal domain (NTD) of Hsp90, which contains a highly conserved ATP-binding pocket essential for the chaperone's function.[2] The drug binds to this pocket with very high affinity, competitively inhibiting the hydrolysis of ATP.[19] This inhibition is highly potent, with half-maximal inhibitory concentrations (
IC50) in cell-free assays measured at 13 nM for the Hsp90α isoform and 21 nM for the Hsp90β isoform.[19] Its activity against other Hsp90 family members, such as the endoplasmic reticulum-resident GRP94 and the mitochondrial TRAP-1, is significantly weaker, indicating a degree of selectivity for the cytosolic isoforms.[23]
A key mechanistic event that serves as a direct readout of target engagement is the disruption of the Hsp90 multichaperone complex. The binding of the co-chaperone p23 to Hsp90 is an ATP-dependent step required for client protein maturation and stability.[35] Luminespib potently and rapidly induces the dissociation of the Hsp90-p23 complex. In cellular assays, this effect occurs within minutes of exposure and at concentrations significantly lower than those required for the first-generation inhibitor 17-AAG, demonstrating Luminespib's superior biochemical potency.[3]
By arresting the Hsp90 chaperone cycle, Luminespib prevents the proper folding and stabilization of client proteins. These destabilized clients are recognized by the cellular quality control machinery and targeted for degradation via the ubiquitin-proteasome pathway.[2] This leads to the rapid and sustained depletion of a wide array of oncoproteins that are critical drivers of malignancy.
Preclinical studies have extensively documented the degradation of key Hsp90 clients following Luminespib treatment, including:
The simultaneous degradation of these multiple, diverse client proteins is the central tenet of Hsp90 inhibition's therapeutic potential. It allows for a pleiotropic attack on the cancer cell, blocking numerous critical signaling pathways at once. This mechanism provides a strong rationale for its use in tumors driven by specific Hsp90-dependent oncoproteins and suggests a potential to overcome acquired resistance to therapies that target a single pathway. For instance, in models of NSCLC with intrinsic resistance to MEK inhibitors, bypass signaling often occurs through PI3K/AKT activation. Because Luminespib degrades clients in both the RAF-MEK-ERK and PI3K-AKT pathways, it was shown preclinically to sensitize resistant cells to MEK inhibition.[36]
The molecular signature of Hsp90 inhibition by Luminespib is consistent and measurable, providing robust pharmacodynamic (PD) biomarkers for assessing target engagement in both preclinical models and clinical trials. The two canonical PD markers are the depletion of sensitive client proteins (e.g., HER2, AKT) and the compensatory induction of other heat shock proteins, most notably Hsp72 (also known as HSPA1A).[3] The upregulation of Hsp72 is a result of the activation of Heat Shock Factor 1 (HSF1) in response to the accumulation of unfolded proteins, and its induction serves as a reliable and dose-dependent indicator that the Hsp90 chaperone has been successfully inhibited.[18]
Luminespib demonstrated potent and broad-spectrum antitumor activity in a comprehensive suite of preclinical models.
Table 2: Summary of Preclinical Activity (GI50/IC50 Values) in Key Cancer Cell Lines | |||||
---|---|---|---|---|---|
Cell Line | Cancer Type | Key Genetic Feature | Assay Type | Value (nM) | Source |
A2780 | Ovarian Cancer | - | IC50 | 1 | 31 |
BT-474 | Breast Cancer | HER2+ | GI50 | 3.1 | 30 |
BT-474 | Breast Cancer | HER2+ | IC50 | 14 | 31 |
DU-145 | Prostate Cancer | - | GI50 | 5 | 31 |
A549 | NSCLC | KRAS mutant | GI50 | 10-26 | 31 |
EBC-1 | NSCLC | MET amplified | IC50 | 13 | 31 |
NCI-N87 | Gastric Cancer | HER2+ | GI50 | ~2-40 | 19 |
SNU-216 | Gastric Cancer | - | GI50 | ~2-40 | 19 |
U87MG | Glioblastoma | PTEN null | - | Potent (nM range) | 12 |
The robust preclinical profile of Luminespib provided a strong rationale for its advancement into human clinical trials. The program, managed by Novartis, was extensive, exploring the drug as a single agent and in combination across a range of advanced solid tumors, with a particular focus on molecularly defined patient populations.
Table 3: Overview of Major Clinical Trials for Luminespib (AUY922) | ||||||
---|---|---|---|---|---|---|
Trial Identifier | Phase | Indication(s) | Patient Population | Drug(s) | Status | Source |
NCT00526045 | I/II | Advanced Solid Malignancies; Breast Cancer | HER2+ or ER+ advanced/metastatic | AUY922 | Completed | 38 |
Not Specified | I | Advanced Solid Malignancies | General population | AUY922 | Completed | 37 |
NCT01124864 | II | Non-Small Cell Lung Cancer (NSCLC) | Previously treated, molecularly defined | AUY922 | Completed | 39 |
NCT01752400 | II | Non-Small Cell Lung Cancer (NSCLC) | Advanced ALK-positive | AUY922 | Completed | 39 |
NCT01854034 | II | Non-Small Cell Lung Cancer (NSCLC) | EGFR Exon 20 insertion mutations | AUY922 | Completed | 39 |
NCT01294826 | I | Colorectal Cancer | KRAS wild-type metastatic | AUY922 + Cetuximab | Completed | 24 |
NCT01402401 | II | Gastric Cancer | Second-line HER2-positive | AUY922 + Trastuzumab | Terminated | 41 |
The initial clinical evaluation of Luminespib occurred in first-in-human, Phase I dose-escalation studies designed to establish its safety, tolerability, and pharmacokinetic (PK) and pharmacodynamic (PD) profiles.[37] In a key study, 96 to 101 patients with various advanced solid tumors received weekly one-hour intravenous infusions of AUY922 at doses escalating from 2 mg/m² to 70 mg/m².[37]
The study successfully established the Maximum Tolerated Dose (MTD) at 70 mg/m² once weekly, which was subsequently selected as the Recommended Phase II Dose (RP2D).[37] PK analysis revealed that Luminespib's blood concentration followed a bi-exponential decline, characterized by a rapid initial distribution phase (half-life
<10 minutes) and a prolonged terminal elimination phase (half-life ≈60 hours), supporting the weekly dosing schedule.[37] PD assessments confirmed dose-dependent target engagement, as evidenced by the induction of the biomarker Hsp72 in peripheral blood mononuclear cells.[37] While objective tumor responses were not a primary endpoint, preliminary signs of activity were observed, with disease stabilization reported in 16 patients and partial metabolic responses on FDG-PET scans in nine patients.[37]
The investigation of Luminespib in NSCLC was a cornerstone of its clinical program, driven by the strong dependence of key NSCLC oncogenes, such as mutant EGFR and ALK fusion proteins, on Hsp90. A large, multi-cohort Phase II study (NCT01124864) enrolled 153 heavily pretreated patients with advanced NSCLC, who were stratified into cohorts based on their tumor's molecular status (ALK-rearranged, EGFR-mutant, KRAS-mutant, or wild-type for all three).[39]
The results from this study powerfully illustrated the concept of oncogene addiction as a predictor of sensitivity to Hsp90 inhibition.
Given the established role of Hsp90 in stabilizing both the estrogen receptor (ER) and the HER2 oncoprotein, breast cancer was another logical indication for Luminespib. A Phase I/II study (NCT00526045) included an expansion cohort for patients with ER-positive or HER2-positive advanced or metastatic breast cancer.[38] This part of the trial was terminated early, but of the 16 women enrolled, one achieved a partial response and another had stable disease, providing a modest signal of activity.[45]
More promising results emerged from a Phase Ib/II trial that evaluated Luminespib in combination with the anti-HER2 antibody trastuzumab in patients with HER2-positive metastatic breast cancer who had progressed on prior trastuzumab-based therapies.[16] In the cohort of 41 patients treated at the RP2D (70 mg/m² AUY922 plus standard trastuzumab), the combination yielded an ORR of 22.0% and a stable disease rate of 48.8%.[16] This demonstrated that inhibiting Hsp90 could re-sensitize tumors to HER2-targeted therapy, likely by promoting the degradation of the HER2 receptor.
Luminespib was also evaluated in gastrointestinal cancers. A Phase I study (NCT01294826) explored its combination with cetuximab in KRAS wild-type metastatic colorectal cancer, though specific efficacy outcomes are not detailed in the available materials.[24] A Phase II trial (NCT01402401) was initiated to study Luminespib plus trastuzumab in second-line HER2-positive gastric cancer, but this trial was terminated, potentially due to lack of efficacy, toxicity, or a strategic portfolio decision by the sponsor.[41]
The clinical development of Luminespib provided a comprehensive understanding of its safety profile. While it successfully avoided the hepatotoxicity of first-generation inhibitors, its high potency was associated with a distinct and challenging set of on-target adverse events that ultimately limited its therapeutic potential.
Across all clinical trials, a consistent pattern of treatment-related adverse events (AEs) was observed. The most frequently reported toxicities were gastrointestinal and constitutional.[37] Diarrhea was the most common AE, reported in 55% to over 70% of patients in various studies, followed by nausea (approx. 35%), fatigue or asthenia (approx. 32%), and decreased appetite.[37] While most of these events were Grade 1 or 2 in severity, they could be persistent and impact quality of life.
The MTD of 70 mg/m² weekly was defined in Phase I studies by the occurrence of Grade 3 dose-limiting toxicities (DLTs). These included severe diarrhea, asthenia/fatigue, and anorexia.[37] A Grade 3 atrial flutter was also reported at a lower dose (22 mg/m²), indicating a potential for cardiac effects.[37] The combination of these toxicities established a clear ceiling for safe dose escalation.
The most distinctive and clinically significant toxicity associated with Luminespib was ocular. A high percentage of patients, up to nearly 80% in the Phase II NSCLC study, experienced visual-related disorders.[16] The characteristic symptoms included night blindness (nyctalopia), reported by approximately 20-23% of patients, blurred vision, photopsia (perceptions of flashing lights), and delayed accommodation to changes in light.[37]
This ocular toxicity is considered an on-target effect, likely related to the essential role of Hsp90 in maintaining the structural and functional integrity of proteins in retinal photoreceptor cells, which have extremely high metabolic and protein turnover rates. While the majority of these events were Grade 1-2 and were reported to be reversible upon discontinuation of the drug, they were dose-dependent and became more frequent and severe at doses of 40 mg/m² and above.[15] Crucially, Grade 3 visual symptoms, such as darkening of vision, were observed at the 70 mg/m² dose level and constituted a DLT, leading to dose interruptions, reductions, and in some cases, permanent discontinuation of treatment.[16] This unavoidable on-target toxicity in a critical sensory organ represented a major liability for the drug and a significant challenge for the entire class of pan-Hsp90 inhibitors.
Table 4: Summary of Common and Dose-Limiting Adverse Events (AEs) | |||
---|---|---|---|
Adverse Event | Reported Frequency (All Grades) | Common Grade | Grade ≥3 / DLT Frequency |
Diarrhea | 55-77% 37 | 1-2 | DLT at 40, 54, and 70 mg/m² 37 |
Ocular Toxicities (e.g., Night Blindness, Blurred Vision) | 20-80% 37 | 1-2 | DLT (darkening of vision) at 70 mg/m² 37 |
Fatigue / Asthenia | 32-65% 37 | 1-2 | DLT at 40 and 54 mg/m² 37 |
Nausea | 35% 37 | 1-2 | Uncommon |
Vomiting | 19% 37 | 1-2 | Uncommon |
Anorexia (Decreased Appetite) | ~40% 40 | 1-2 | DLT at 40 mg/m² 37 |
Atrial Flutter | Infrequent | - | DLT at 22 mg/m² 37 |
The trajectory of Luminespib from a promising preclinical candidate to its eventual discontinuation provides a salient case study in modern drug development, reflecting the interplay of scientific discovery, clinical trial outcomes, and strategic corporate decision-making.
Luminespib was the product of a successful academic-industry collaboration between The Institute of Cancer Research (ICR) and the UK-based pharmaceutical company Vernalis.[11] Using structure-based drug design, they developed AUY922 as a potent, synthetic Hsp90 inhibitor. Recognizing its potential, Novartis licensed the program from Vernalis in 2004 to lead its global clinical development.[48]
Under Novartis's stewardship, Luminespib entered Phase I clinical trials in 2007, with Phase II studies commencing in 2011.[8] The drug's potent mechanism and promising early data led to significant optimism within Novartis. In 2011, the company highlighted AUY922 in its pipeline presentation as one of seven potential "blockbuster" assets, with a projected regulatory submission in 2015.[49] This designation underscored the high expectations for Hsp90 inhibition as a major new therapeutic modality in oncology.
Despite the initial optimism, Novartis announced in December 2014 that it had ceased all development work on AUY922.[48] The rights to the program subsequently reverted to Vernalis. The publicly stated reason for the discontinuation was the failure of the drug to meet its primary endpoint of complete and partial response in a Phase II clinical trial in NSCLC patients.[51]
While failure to meet a primary endpoint is a clear trigger for halting development, a more nuanced analysis suggests the decision was likely driven by a confluence of factors that collectively diminished the drug's projected value and strategic fit:
The comprehensive evaluation of Luminespib (AUY922) offers critical lessons on the translation of a potent biological mechanism into a viable therapeutic. Its story is not one of simple failure, but rather a detailed illustration of the complex interplay between molecular potency, on-target toxicity, tumor biology, and the strategic realities of pharmaceutical development.
Luminespib can be regarded as a pinnacle of second-generation Hsp90 inhibitor design. It successfully addressed the significant pharmaceutical and metabolic liabilities of its ansamycin predecessors, delivering a potent, soluble, and synthetically accessible molecule. The clinical program unequivocally validated Hsp90 as a druggable target in human cancer. The clear pharmacodynamic evidence of Hsp72 induction following treatment demonstrated robust and consistent target engagement, confirming that the drug was performing its intended biochemical function in vivo.[37] However, the program also revealed the inherent flaw in the pan-Hsp90 inhibitor strategy: the potency that made it effective against cancer cells was inseparable from the toxicity it caused in sensitive normal tissues. The high incidence of ocular and gastrointestinal AEs is a direct consequence of inhibiting a chaperone that is vital for cellular homeostasis in high-turnover tissues. This "potency-toxicity paradox" created a narrow therapeutic window that ultimately limited the drug's clinical utility.
The journey of Luminespib provides several enduring lessons for oncology drug development:
While the development of Luminespib has been terminated, the knowledge gained from its extensive investigation continues to guide the field. The challenges encountered with Luminespib and other pan-Hsp90 inhibitors have catalyzed a shift towards more sophisticated strategies. Current research focuses on developing isoform-selective inhibitors (e.g., targeting Hsp90β to potentially spare retinal tissue), inhibitors that target the C-terminal domain or co-chaperone interactions, and proteolysis-targeting chimera (PROTAC) approaches to selectively degrade Hsp90 in tumor cells.[8] Pimitespib (TAS-116), an oral Hsp90 inhibitor with a potentially more manageable safety profile, has recently gained approval in Japan for gastrointestinal stromal tumors, suggesting that the therapeutic potential of Hsp90 inhibition can be realized with the right molecule and indication.[53]
In conclusion, Luminespib (AUY922) stands as a landmark compound in the history of Hsp90-targeted therapy. It was a potent and well-characterized research tool that provided definitive proof-of-concept for Hsp90 inhibition in humans. Its ultimate failure to reach the market was not due to a lack of potency or a flawed mechanism, but rather to the inherent biological complexities of its target, which serve as a crucial and enduring lesson for the future development of drugs targeting fundamental cellular machinery.
Published at: September 5, 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.