MedPath

Tasquinimod Advanced Drug Monograph

Published:Sep 22, 2025

Generic Name

Tasquinimod

Drug Type

Small Molecule

Chemical Formula

C20H17F3N2O4

CAS Number

254964-60-8

Tasquinimod (DB05861): A Comprehensive Report on a Pleiotropic TME-Modulating Agent from Prostate Cancer Failure to Hematological Repurposing

1.0 Executive Summary

Tasquinimod (DB05861) is an orally active, investigational small molecule belonging to the quinoline-3-carboxamide class. It represents a second-generation analogue developed from the immunomodulatory agent roquinimex, engineered to possess a more favorable therapeutic index. The compound is distinguished by a novel, pleiotropic mechanism of action that does not directly target cancer cells but instead modulates the tumor microenvironment (TME) through dual inhibition of key pathological pathways. This unique mode of action, which encompasses immunomodulatory, anti-angiogenic, and anti-metastatic properties, has positioned Tasquinimod as a subject of significant clinical interest and strategic re-evaluation over its development history.

The clinical development of Tasquinimod is a story of stark contrast. Initially developed for solid tumors, it demonstrated considerable promise in a large, randomized Phase II trial for metastatic castration-resistant prostate cancer (mCRPC), where it significantly prolonged progression-free survival (PFS) compared to placebo.[1] These compelling results prompted a broad partnership between Active Biotech and Ipsen and led to the initiation of a large-scale, global Phase III trial (10TASQ10) designed to confirm its efficacy and establish an overall survival (OS) benefit.[3] The trial successfully met its primary endpoint, once again demonstrating a statistically significant improvement in radiographic PFS (rPFS). However, it critically failed to confer any benefit in OS, the key secondary endpoint, ultimately revealing an unfavorable risk-benefit profile in this patient population. This outcome led to the discontinuation of the entire prostate cancer development program in 2015.[5]

Despite this significant setback, the deep scientific understanding of Tasquinimod's mechanism of action provided a clear rationale for its strategic repurposing. Research has established that the drug's effects are mediated primarily through the binding and inhibition of the S100A9 protein and the allosteric modulation of Histone Deacetylase 4 (HDAC4).[7] The S100A9 pathway is particularly critical in the bone marrow microenvironment, a key pathological niche in hematological malignancies. Consequently, development has been strategically pivoted towards myelofibrosis (MF) and multiple myeloma (MM), where preclinical models have shown significant therapeutic potential.[8]

Currently, Tasquinimod is being investigated in Phase II clinical trials for MF, both as a monotherapy and in combination with JAK inhibitors, and has completed a Phase Ib/IIa study in heavily pretreated MM, where it demonstrated clinical activity and the potential to overcome therapeutic resistance.[10] Its potential in these rare diseases has been recognized by the U.S. Food and Drug Administration (FDA), which has granted it Orphan Drug Designations for both MM and MF.[12] Tasquinimod's journey serves as a compelling case study in pharmaceutical development, illustrating the fallibility of surrogate endpoints in oncology, the resilience of a scientifically well-understood asset, and the strategic imperative of mechanism-driven drug repurposing.

2.0 Compound Profile and Physicochemical Properties

A comprehensive understanding of Tasquinimod begins with its fundamental chemical and physical characteristics, which define its identity, classification, and behavior in biological systems. These properties form the basis for its formulation as an orally active agent and predict its pharmacokinetic profile.

2.1 Identification and Nomenclature

Tasquinimod is known by several names and unique identifiers across chemical, regulatory, and research databases, ensuring its unambiguous identification.

  • Generic Name: Tasquinimod [14]
  • Synonyms: Common synonyms used during its development and in scientific literature include ABR-215050, TASQ, and ABR-5050.[15]
  • DrugBank ID: DB05861 [14]
  • CAS Number: The Chemical Abstracts Service registry number is 254964-60-8.[14]
  • IUPAC Name: Its systematic chemical name is 4-hydroxy-5-methoxy-N,1-dimethyl-2-oxo-N-[4-(trifluoromethyl)phenyl]-1,2-dihydroquinoline-3-carboxamide.[14]

2.2 Chemical Structure and Classification

Tasquinimod's molecular structure and chemical class are central to its biological activity.

  • Chemical Formula: The empirical formula for Tasquinimod is C20​H17​F3​N2​O4​.[14]
  • Molecular Weight: The average molecular weight is 406.361 g/mol, with a monoisotopic mass of 406.114041524 Da.[14]
  • Structural Identifiers: For computational and database purposes, it is defined by the following identifiers:
  • SMILES: CN1C2=C(C(=CC=C2)OC)C(=C(C1=O)C(=O)N(C)C3=CC=C(C=C3)C(F)(F)F)O [15]
  • InChIKey: ONDYALNGTUAJDX-UHFFFAOYSA-N [15]
  • Chemical Class: Tasquinimod is a synthetic organic compound classified as a quinoline-3-carboxamide. It was developed as a second-generation analogue of roquinimex, retaining the core quinoline scaffold but modified for enhanced potency and a different safety profile.[15] Chemically, it belongs to the class of aromatic anilides, characterized by a carboxamide group substituted with an aromatic ring.[14] Its structure incorporates a dihydroquinoline core, an anisole (methoxybenzene) group, and a trifluoromethylphenyl moiety, which contribute to its overall lipophilicity and binding interactions.[14]

2.3 Physicochemical and Predicted ADME Characteristics

The physicochemical properties of Tasquinimod are highly indicative of a molecule designed for oral administration and good membrane permeability. These characteristics, summarized in Table 1, directly inform its clinical pharmacology.

  • Solubility and Lipophilicity: The compound exhibits very low aqueous solubility at 0.00911 mg/mL, a common feature of orally absorbed drugs that follow dissolution-limited absorption.[14] It is, however, readily soluble in organic solvents such as dimethyl sulfoxide (DMSO) and dimethylformamide (DMF).[20] Its significant lipophilicity is quantified by its partition coefficient (logP), with calculated values of 3.26 and 2.31, suggesting a high propensity to cross biological membranes.[14]
  • Medicinal Chemistry Properties: Tasquinimod's molecular structure aligns well with established guidelines for "drug-likeness." It satisfies Lipinski's Rule of Five and the Ghose Filter, both of which are predictors of good oral bioavailability.[14] The molecule has one hydrogen bond donor and four hydrogen bond acceptors. It fails Veber's Rule, which is likely attributable to its four rotatable bonds, suggesting a degree of conformational flexibility that may impact permeability but does not preclude oral absorption.[14]
  • Predicted ADME Profile: In silico modeling provides further insight into its likely behavior in the human body. These models predict a very high probability of human intestinal absorption (0.9941) and an ability to cross the blood-brain barrier (0.991).[14] Furthermore, Tasquinimod is predicted to be neither a substrate nor an inhibitor of the P-glycoprotein (P-gp) efflux pump, which reduces the risk of certain drug-drug interactions and mechanisms of resistance.[14] Computational toxicology predictions are also favorable, suggesting the compound is non-mutagenic (Non-AMES toxic) and non-carcinogenic, with low potential for promiscuous inhibition of cytochrome P450 enzymes.[14]

The combination of these fundamental chemical characteristics provides a clear and direct explanation for one of Tasquinimod's most critical clinical features: its efficacy as an orally active agent. The high lipophilicity (logP > 2.3), coupled with structural properties that align with the Rule of Five, results in the high predicted intestinal absorption that is borne out in clinical practice.[7] This allows for a convenient once-daily oral dosing regimen, a significant advantage for chronic cancer therapy, and was a foundational element of its development strategy from the outset.[23]

Table 1: Key Identifiers and Physicochemical Properties of Tasquinimod

PropertyValueSource(s)
Identifiers
DrugBank IDDB0586114
CAS Number254964-60-814
IUPAC Name4-hydroxy-5-methoxy-N,1-dimethyl-2-oxo-N-[4-(trifluoromethyl)phenyl]-1,2-dihydroquinoline-3-carboxamide14
Chemical Formula & Weight
Chemical FormulaC20​H17​F3​N2​O4​14
Average Molecular Weight406.361 g/mol14
Monoisotopic Mass406.114041524 Da14
Physicochemical Properties
Water Solubility0.00911 mg/mL14
logP (Partition Coefficient)3.26 (ALOGPS), 2.31 (Chemaxon)14
pKa (Strongest Acidic)4.7314
Polar Surface Area70.08 Ų14
Rotatable Bond Count414
Medicinal Chemistry Rules
Rule of FiveYes14
Ghose FilterYes14
Veber's RuleNo14
Predicted Properties
Bioavailability1 (Chemaxon Prediction)14
Human Intestinal Absorption+ (0.9941 Probability)14
Blood Brain Barrier+ (0.991 Probability)14

3.0 Preclinical Pharmacology: A Dual-Targeting Mechanism of Action

Tasquinimod is characterized by a pleiotropic mechanism of action that is unconventional among oncology agents. Rather than inducing direct cytotoxicity in tumor cells, it modulates the complex ecosystem of the TME. Its anti-cancer effects arise from simultaneously targeting two distinct but interconnected proteins: the extracellular inflammatory protein S100A9 and the intracellular epigenetic regulator HDAC4. This dual-targeting strategy disrupts the reciprocal signaling pathways that cancer cells use to co-opt host cells for survival, proliferation, and metastasis.

3.1 Targeting the S100A9-TLR4/RAGE Axis

The primary extracellular mechanism of Tasquinimod involves its high-affinity binding to S100A9, a calcium- and zinc-binding protein also known as Calgranulin B or migration inhibitory factor-related protein 14 (MRP-14).[7] S100A9 is a critical damage-associated molecular pattern (DAMP) molecule, typically expressed and secreted by myeloid cells such as neutrophils and monocytes, particularly under conditions of inflammation or stress within the TME.[25]

Once secreted, S100A9 acts as a potent signaling molecule, promoting tumor progression through several mechanisms. It interacts with pattern recognition receptors on the surface of other immune cells, most notably Toll-like receptor 4 (TLR4) and the receptor for advanced glycation end products (RAGE).[7] This engagement triggers pro-inflammatory and pro-tumorigenic signaling cascades that are essential for the recruitment, accumulation, and activation of immunosuppressive myeloid cell populations, including myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs).[25] These cells, in turn, create an environment that fosters angiogenesis and immune evasion.[25]

Tasquinimod directly intervenes in this process. It binds to S100A9 in a zinc-dependent manner, physically obstructing its interaction with TLR4 and RAGE.[7] By blocking this critical signaling axis, Tasquinimod effectively cuts off a key pathway that tumors use to manipulate the host immune system for their benefit.

3.2 Allosteric Modulation of HDAC4

Complementing its extracellular activity, Tasquinimod exerts a profound intracellular effect through its interaction with Histone Deacetylase 4 (HDAC4), a class IIa histone deacetylase.[7] Unlike pan-HDAC inhibitors, Tasquinimod's action is highly specific and does not involve direct inhibition of the enzyme's catalytic site. Instead, it functions as an allosteric modulator.[18]

Tasquinimod binds with high affinity (dissociation constant, Kd​, of 10–30 nM) to the regulatory zinc-binding domain of HDAC4.[30] This binding event induces a conformational change that "locks" the HDAC4 protein in an inactive state.[29] The critical consequence of this conformational lock is the prevention of the assembly of the HDAC4/N-CoR/HDAC3 transcriptional repressor complex.[7] This multi-protein complex is essential for the deacetylation of histone and non-histone proteins, which leads to epigenetic silencing of target genes.

By preventing the formation of this repressor complex, Tasquinimod effectively inhibits the deacetylation of key HDAC4 client proteins. One of the most important of these is Hypoxia-Inducible Factor 1-alpha (HIF-1α), a master transcription factor that orchestrates the cellular response to low oxygen levels (hypoxia)—a hallmark of the TME.[20] Under hypoxic conditions, HIF-1α drives the expression of genes involved in angiogenesis, cell survival, and metastasis. Tasquinimod's inhibition of HDAC4-mediated HIF-1α deacetylation suppresses this adaptive hypoxic response in both cancer and endothelial cells, striking at the heart of the tumor's survival strategy.[19]

3.3 Downstream Pharmacodynamic Effects on the Tumor Microenvironment

The concurrent inhibition of the S100A9 and HDAC4 pathways by Tasquinimod triggers a cascade of favorable downstream effects that collectively re-engineer the TME from a tumor-supportive to a tumor-hostile state.

3.3.1 Immunomodulation

Tasquinimod fundamentally reshapes the immune composition of the tumor. By blocking S100A9 signaling, it curtails the accumulation and immunosuppressive function of MDSCs.[19] Furthermore, it induces a phenotypic shift in the TAM population. It promotes the repolarization of macrophages from the pro-tumor, pro-angiogenic, and immunosuppressive M2 phenotype towards the pro-inflammatory and anti-tumor M1 phenotype.[25] This early change in macrophage polarization is a key mechanism of its anti-tumor action, as it reduces local immune suppression and can render the tumor more susceptible to other forms of immunotherapy. Indeed, preclinical studies have shown that the immunomodulatory effects of Tasquinimod can enhance the anti-tumor activity of PD-L1 blockade.[28]

3.3.2 Anti-Angiogenesis

The drug's anti-angiogenic effects are multi-faceted and not dependent on the direct inhibition of VEGF or its receptors.[19] The primary mechanism is the suppression of the HIF-1α hypoxic response via HDAC4 modulation, which leads to the downregulation of critical pro-angiogenic genes, including VEGF.[20] This is complemented by a distinct mechanism: Tasquinimod has been shown to directly upregulate the expression of Thrombospondin-1 (TSP-1), a potent endogenous inhibitor of angiogenesis.[20] This dual action—suppressing pro-angiogenic drivers while simultaneously promoting an anti-angiogenic factor—results in a robust inhibition of new blood vessel formation within the tumor.

3.3.3 Anti-Metastatic Activity

Tasquinimod's ability to inhibit metastasis stems from its impact on both immune cells and the formation of receptive sites for tumor spread. The inhibition of S100A9 is particularly critical, as this protein is known to play a role in establishing pre-metastatic niches—microenvironments in distant organs that are primed to support the growth of disseminated tumor cells.[19] By disrupting S100A9 signaling, Tasquinimod interferes with this preparatory process. This, combined with its broad anti-angiogenic and immunomodulatory effects, creates an environment that is less conducive to both the escape of primary tumor cells and the establishment of secondary tumors.[17]

The dual mechanism of action of Tasquinimod is not merely additive; it is synergistic, as it targets a self-perpetuating pathological feedback loop within the TME. The process begins with tumor-induced hypoxia, which stabilizes and activates the transcription factor HIF-1α.[25] This activation drives the expression of genes that promote tumor survival and angiogenesis. Critically, HIF-1α also enhances the transcription and secretion of S100A9 by both cancer and myeloid cells.[7] The secreted S100A9 then acts as a chemoattractant, recruiting more immunosuppressive MDSCs to the tumor site.[7] These MDSCs further contribute to the hypoxic and immunosuppressive TME, thereby reinforcing the initial stimulus and completing a "vicious cycle" that sustains tumor growth. Tasquinimod disrupts this cycle at two distinct, critical nodes. First, by allosterically modulating HDAC4, it prevents HIF-1α activation, thus reducing the primary stimulus for S100A9 production.[29] Second, by directly binding to and inhibiting extracellular S100A9, it blocks the recruitment of MDSCs, preventing the reinforcement of the cycle.[25] This simultaneous attack on both the instigating signal (HIF-1α) and the reinforcing signal (S100A9) leads to a collapse of the tumor-supportive microenvironment, an effect more profound than could be achieved by targeting either pathway in isolation. This intricate interplay explains its characterization as a pleiotropic agent with broad effects on the TME.[25]

4.0 Clinical Pharmacology: Pharmacokinetics and Distribution

The clinical utility of Tasquinimod is underpinned by a favorable pharmacokinetic (PK) profile characterized by excellent oral absorption, a long half-life suitable for once-daily dosing, and a unique distribution mechanism that concentrates the drug within the TME.

4.1 Absorption, Distribution, Metabolism, and Excretion (ADME)

The ADME properties of Tasquinimod have been well-defined through Phase I and II clinical trials.

  • Absorption: Consistent with its physicochemical properties, Tasquinimod demonstrates excellent oral bioavailability.[7] Following oral administration, maximum plasma concentrations ( Cmax​) are achieved in approximately 2.6 hours (Tmax​).[24] Clinical studies have shown that its absorption and overall PK profile are not significantly affected by co-administration with food, allowing for flexible dosing without dietary restrictions.[24]
  • Distribution: Tasquinimod has a relatively small volume of distribution of 5.9 L, indicating that it does not distribute extensively into peripheral tissues outside of the plasma compartment.[24] This is a direct consequence of its high degree of plasma protein binding, which exceeds 98%.[7] This binding is almost exclusively to serum albumin, the most abundant protein in plasma.[7]
  • Metabolism: The primary route of metabolism for Tasquinimod is hepatic, mediated by the cytochrome P450 enzyme CYP3A4.[7] While it is converted to several metabolites, preclinical studies have definitively shown that the parent compound is the active therapeutic moiety. Experiments using ketoconazole, a potent inhibitor of CYP3A4, demonstrated that blocking Tasquinimod's metabolism did not diminish its anti-cancer efficacy in vivo, confirming that the pharmacological activity resides with the parent drug and not its metabolites.[7]
  • Excretion: Tasquinimod is characterized by low systemic clearance, with values of 0.19 L/h at a 0.5 mg dose and 0.22 L/h at a 1 mg dose.[24] This low clearance contributes to its long elimination half-life, which is approximately 40 ± 16 hours.[7] This extended half-life is highly advantageous, as it supports a convenient and compliance-friendly once-daily oral dosing schedule. Pharmacokinetic analyses have also revealed that clearance decreases slightly with age (approximately 1.4% per year), which can lead to increased drug exposure in elderly patients. This finding has clinical relevance, as dose reductions were often required for patients over 80 years of age to manage toxicities associated with higher exposure.[35]

4.2 The Role of Albumin Binding and the EPR Effect

A critical and sophisticated aspect of Tasquinimod's pharmacology is its interaction with albumin, which facilitates a form of passive tumor targeting via the Enhanced Permeability and Retention (EPR) effect. This mechanism is key to understanding its therapeutic potency in vivo.

Tasquinimod binds reversibly but with high affinity (dissociation constant, Kd​<35 μM) to the IIA subdomain of serum albumin, also known as Sudlow's site I.[7] Due to the high concentration of albumin in the blood, over 98% of circulating Tasquinimod is bound to this protein at any given time.[7]

The tumor microenvironment is characterized by abnormal, hastily constructed blood vessels that are significantly "leakier" than those in healthy tissues. These leaky vessels have poorly formed endothelial junctions, allowing large molecules like albumin and albumin-bound drugs to extravasate from the bloodstream into the tumor's interstitial space. Coupled with poor lymphatic drainage from the tumor, this leads to the accumulation of these macromolecules at the tumor site. This phenomenon is the EPR effect.[7]

The EPR effect provides a compelling explanation for a key pharmacological paradox observed with Tasquinimod. In standard in vitro cell culture assays, the concentration of Tasquinimod required to inhibit cancer cell growth by 50% (IC50​) is approximately 50 μM.[38] However, in clinical trials, therapeutic effects are observed with chronic daily dosing that maintains steady-state plasma concentrations of only ~0.5 μM—a level 100-fold lower than the in vitro

IC50​.[7] The EPR effect reconciles this discrepancy. The passive accumulation of the albumin-Tasquinimod complex within the TME concentrates the drug at its site of action. This results in intracellular drug concentrations within the tumor reaching levels of 2–3 μM.[7] These locally elevated concentrations are several-fold higher than the 0.5 μM required to inhibit endothelial sprouting and are well within the range needed to effectively inhibit its molecular targets, HDAC4 and S100A9. This mechanism essentially acts as a passive targeting system, enhancing the drug's therapeutic index by maximizing its concentration in the tumor while keeping systemic free-drug levels low, which may help mitigate systemic toxicity.

5.0 Clinical Development in Prostate Cancer: A Dichotomy of Efficacy

The clinical development of Tasquinimod for metastatic castration-resistant prostate cancer (mCRPC) is a compelling narrative of initial promise followed by pivotal failure. This journey provides critical insights into the challenges of drug development in modern oncology, particularly concerning the selection of clinical trial endpoints.

5.1 Phase II Trial (NCT00560482): A Foundation of Promise

The foundation for Tasquinimod's large-scale development was built upon the highly successful results of a randomized, double-blind, placebo-controlled Phase II trial.[2] The study enrolled 201 men with minimally symptomatic, chemotherapy-naïve mCRPC and was designed to assess the drug's ability to delay disease progression.[2]

The trial met its primary endpoint with a high degree of statistical significance. The proportion of patients who remained progression-free at 6 months was 69% in the Tasquinimod arm, compared to just 37% in the placebo arm (P<.001).[2] This translated into a more than doubling of the median progression-free survival (PFS), which was 7.6 months for patients receiving Tasquinimod versus 3.3 months for those on placebo (Hazard Ratio 0.57;

P=.0042).[2] This robust PFS benefit was consistently observed across all clinically relevant subgroups, including patients with nodal, bone, and visceral metastases, suggesting a broad applicability within the mCRPC population.[2]

Furthermore, long-term follow-up data from this study was encouraging. While not statistically powered for overall survival, exploratory analyses suggested a potential OS advantage, with a median OS of 34.2 months in the Tasquinimod group versus 30.2 months in the placebo group.[3] This signal, particularly strong in the subgroup of patients with bone metastases, provided a compelling rationale to advance Tasquinimod into a definitive Phase III trial powered to confirm an OS benefit.[3]

5.2 The Pivotal Phase III Trial (10TASQ10 / NCT01234311): Success and Failure

Based on the strength of the Phase II data, Active Biotech and its partner Ipsen launched the 10TASQ10 study, a large, global, pivotal Phase III trial.[3]

5.2.1 Study Design and Endpoints

The trial was a multinational, randomized (2:1), double-blind, placebo-controlled study that enrolled 1,245 men with chemotherapy-naïve mCRPC who had bone metastases.[3] The study's design mirrored the successful Phase II trial but on a much larger scale, with sufficient statistical power for definitive conclusions. The primary endpoint was radiographic progression-free survival (rPFS), as determined by central, blinded review. The key secondary endpoint, and the ultimate measure of clinical benefit required for regulatory approval, was overall survival (OS).[3]

5.2.2 Efficacy Outcomes

The 10TASQ10 trial produced a dichotomous and ultimately disappointing result. It successfully met its primary endpoint, confirming the findings from the Phase II study. Tasquinimod demonstrated a statistically significant improvement in rPFS, with a median of 7.0 months compared to 4.4 months for placebo (HR 0.64; P<.001).[3] This result unequivocally showed that Tasquinimod is an active agent that can slow the radiographic progression of mCRPC.

However, this success was completely overshadowed by the failure to meet the key secondary endpoint. The trial demonstrated no overall survival benefit for Tasquinimod. In fact, the results trended in the wrong direction, with a median OS of 21.3 months in the Tasquinimod arm versus 24.0 months in the placebo arm (HR 1.10; P=.25).[3] The improvement in rPFS did not translate into patients living longer, which is the gold standard for a successful cancer therapy.

5.3 Discontinuation and Lessons Learned

The unambiguous failure to demonstrate an OS benefit, coupled with a greater incidence of adverse events in the treatment arm, led to an unfavorable risk-benefit assessment. On April 16, 2015, Active Biotech and Ipsen announced the formal discontinuation of the Tasquinimod development program for prostate cancer.[6]

The failure of the 10TASQ10 trial, despite meeting its primary endpoint, serves as a prominent case study in the limitations of surrogate endpoints in contemporary oncology. The disconnect between rPFS and OS in this trial is likely not a reflection of a flawed drug mechanism alone, but rather a consequence of the evolving treatment landscape for mCRPC.[5] At the time the trial was conducted, several other life-prolonging therapies (such as abiraterone, enzalutamide, and docetaxel) were available to patients upon progression.[1] Patients in the placebo arm, who progressed radiographically about 2.6 months earlier than those on Tasquinimod, were able to receive these subsequent effective therapies sooner. This access to multiple lines of effective post-progression treatment likely extended their survival, effectively masking or "crossing over" any potential survival benefit conferred by Tasquinimod earlier in the disease course. This phenomenon highlights a critical challenge in clinical trial design: in cancers with multiple effective therapies, a modest improvement in an intermediate endpoint like PFS may not be sufficient to impact the ultimate endpoint of OS. As noted by key opinion leaders at the time, this outcome underscored that rPFS is not a reliable surrogate for OS in mCRPC and that randomized Phase II results can be misleading, prompting calls for more intelligent trial designs and a higher bar for advancing drugs into Phase III studies in this disease space.[5]

Table 2: Summary of Pivotal Clinical Trials in Metastatic Castration-Resistant Prostate Cancer

FeaturePhase II Trial (NCT00560482)Phase III Trial (10TASQ10 / NCT01234311)
Phase23
N (Patients)2011,245
Patient PopulationMinimally symptomatic, chemo-naïve mCRPCAsymptomatic to mildly symptomatic, chemo-naïve mCRPC with bone metastases
Primary EndpointProgression-free proportion at 6 monthsRadiographic Progression-Free Survival (rPFS)
Median PFS (Drug vs. Placebo)7.6 months vs. 3.3 months7.0 months vs. 4.4 months
PFS Hazard Ratio (95% CI)0.57 (0.39, 0.85)0.64 (0.54, 0.75)
Median OS (Drug vs. Placebo)34.2 months vs. 30.2 months (exploratory)21.3 months vs. 24.0 months
OS Hazard Ratio (95% CI)0.72 (0.46, 1.12) (exploratory, adjusted)1.10 (0.94, 1.28)
Source(s)23

6.0 Comprehensive Safety and Tolerability Profile

The safety profile of Tasquinimod has been extensively characterized through a robust clinical development program that included over 1,500 patients, equivalent to more than 650 patient-years of exposure.[8] While generally considered manageable, the cumulative toxicity burden was a significant factor in the risk-benefit assessment that led to the discontinuation of its development in prostate cancer.

6.1 Adverse Events Observed in Clinical Trials

Data from the large Phase II and Phase III trials in mCRPC provide a clear picture of Tasquinimod's safety and tolerability.

  • Common Adverse Events: The most frequently reported treatment-emergent adverse events (TEAEs) were typically mild to moderate (Grade 1-2) in severity. These included gastrointestinal issues such as nausea, constipation, decreased appetite, and flatulence; constitutional symptoms like fatigue and asthenia; and musculoskeletal complaints, including arthralgia (joint pain), myalgia (muscle pain), and pain in the extremities.[2] These side effects were often more pronounced during the initial phase of treatment and tended to become less frequent after the first two months of therapy.[34]
  • Laboratory Abnormalities: A characteristic feature of Tasquinimod treatment was the development of specific, generally asymptomatic, laboratory abnormalities. These included transient elevations in pancreatic enzymes (amylase and lipase) and inflammatory biomarkers such as C-reactive protein (CRP) and fibrinogen.[2] Interestingly, clinical analyses found correlations between these laboratory changes and clinical symptoms; for instance, an increase in CRP was associated with a higher incidence of muscle and joint pain, while an increase in amylase was paradoxically associated with a lower risk of gastrointestinal side effects.[35]
  • Serious and High-Grade Adverse Events: While most side effects were low-grade, Grade 3 or higher adverse events were more common in patients receiving Tasquinimod than in those receiving placebo. In the pivotal Phase III trial, 42.8% of patients in the Tasquinimod arm experienced a Grade ≥3 event, compared to 33.6% in the placebo arm.[3] The most common of these severe events were anemia, fatigue, and cancer pain.[3] Additionally, the Phase II trial noted a higher incidence of deep vein thrombosis in the Tasquinimod arm (4%) compared to the placebo arm (0%).[2] Despite these issues, the drug did not appear to increase cardiovascular risk factors such as hypertension or QTc prolongation.[35]

Table 3: Consolidated Safety Profile - Common Adverse Events from Phase III mCRPC Trial (10TASQ10)

Adverse Event (MedDRA Term)Tasquinimod (n=832) All Grades (%)Tasquinimod (n=832) Grade ≥3 (%)Placebo (n=413) All Grades (%)Placebo (n=413) Grade ≥3 (%)
Gastrointestinal
Nausea27%1%16%<1%
Constipation25%<1%16%<1%
Decreased Appetite20%1%7%<1%
Constitutional
Fatigue29%1%18%<1%
Musculoskeletal
Back Pain24%<1%10%<1%
Pain in Extremity19%<1%6%0%
Arthralgia16%<1%7%<1%
Hematological
Anemia12%3%6%1%
Laboratory
Lipase Increased10%5%<1%0%
Amylase Increased10%1%<1%0%
Data adapted from pivotal Phase III trial publications.3

6.2 Risk-Benefit Analysis in the Context of mCRPC

The decision to halt development in prostate cancer was ultimately based on a comprehensive risk-benefit analysis. The tolerability profile, while deemed manageable from a clinical trial perspective, had tangible consequences for patients. The rate of treatment discontinuation due to adverse events was more than double in the Tasquinimod arm compared to the placebo arm (17.9% vs. 8.8%) in the Phase III study.[3]

This higher toxicity burden likely played a direct role in the failure to achieve an OS benefit. The cumulative impact of chronic, low-grade side effects such as fatigue, nausea, and pain can significantly degrade a patient's quality of life and overall performance status. This is reflected in the Phase III trial data, where symptomatically assessed endpoints, including time to symptomatic progression and quality of life deterioration, actually favored the placebo group.[3] A patient's physical condition at the time of disease progression is a critical determinant of their eligibility for, and ability to tolerate, subsequent lines of life-prolonging therapy. It is therefore highly plausible that while Tasquinimod was effectively slowing radiographic tumor growth (improving rPFS), its associated toxicities were simultaneously causing a decline in patient well-being. This could have rendered patients less fit to receive and benefit from subsequent effective treatments upon progression. In contrast, patients on placebo, while progressing sooner radiographically, may have maintained a better performance status for longer, allowing them to derive greater benefit from later-line therapies. This creates a scenario where a drug can succeed on a surrogate endpoint while failing—or even having a detrimental effect—on the ultimate endpoint of overall survival, a critical lesson from the Tasquinimod program.

7.0 A New Trajectory: Repurposing for Hematological Malignancies

Following the discontinuation of its development in prostate cancer, Tasquinimod has been strategically repurposed for hematological malignancies. This pivot was not an arbitrary search for a new indication but a deliberate, mechanism-driven strategy based on the drug's unique mode of action and the specific pathophysiology of diseases rooted in the bone marrow microenvironment.

7.1 Rationale for Myelofibrosis (MF) and Multiple Myeloma (MM)

The scientific rationale for investigating Tasquinimod in MF and MM is exceptionally strong and centers on its primary target, S100A9. Both MF and MM are cancers where the bone marrow microenvironment is not merely a passive scaffold but an active participant in disease pathogenesis.

In myelofibrosis, the bone marrow becomes progressively fibrotic, disrupting normal blood cell production (hematopoiesis).[8] In multiple myeloma, malignant plasma cells thrive within the bone marrow niche, supported by and interacting with a host of non-malignant cells.[26] In both diseases, myeloid cells, including MDSCs, are key components of this pathological microenvironment. These cells secrete high levels of S100A9, which drives inflammation, supports tumor cell growth, promotes fibrosis, and contributes to therapeutic resistance.[26]

Tasquinimod's ability to bind and inhibit S100A9 makes it an ideal candidate to disrupt these processes. By targeting S100A9, it can directly interfere with the tumor-supportive pathways within the bone marrow, with the potential to reduce fibrosis, restore normal hematopoiesis, and inhibit the growth of malignant cells.[8] This mechanism offers a novel therapeutic approach that is distinct from existing treatments like JAK inhibitors in MF or proteasome inhibitors and immunomodulators in MM, suggesting potential for both monotherapy and combination strategies.

7.2 Recent Preclinical and Clinical Evidence

This strong scientific rationale has been substantiated by a growing body of preclinical and early clinical data.

7.2.1 Myelofibrosis (MF)

  • Preclinical Data: The preclinical evidence for Tasquinimod in MF is compelling. In various mouse models of the disease, treatment with Tasquinimod has been shown to reduce key pathological features, including myeloproliferation, splenomegaly (enlarged spleen), and bone marrow fibrosis.[8] Data presented at the American Society of Hematology (ASH) Annual Meeting in 2024 further strengthened this case, demonstrating that Tasquinimod selectively induces lethality in late-stage MF cells while sparing normal cells.[9] The studies also showed that it reduces the leukemia burden and improves survival in animal models. Critically, these preclinical models revealed synergistic activity when Tasquinimod was combined with standard-of-care JAK inhibitors (like ruxolitinib) or investigational BET inhibitors, providing a strong basis for combination therapy trials.[9]
  • Clinical Trials: Based on this promising preclinical work, several clinical trials have been initiated to evaluate Tasquinimod in MF patients:
  • The TasqForce Trial (HOVON 172 / NCT06605586): This is a Phase Ib/II study being conducted in Europe. It is designed to evaluate the safety and efficacy of Tasquinimod in patients with MF who are refractory to, or intolerant of, prior JAK inhibitor therapy.[52]
  • MD Anderson Cancer Center Trial (NCT06327100 / NCI-2024-02551): This US-based Phase II trial is evaluating Tasquinimod both as a monotherapy and in combination with a stable dose of ruxolitinib in patients with various forms of MF.[11]

7.2.2 Multiple Myeloma (MM)

  • Preclinical Data: In vitro studies have shown that Tasquinimod can directly inhibit the proliferation and colony formation of MM cells. This anti-myeloma effect was associated with the downregulation of c-MYC, a critical oncogene in MM pathogenesis.[26]
  • Clinical Trial (NCT04405167): A Phase Ib/IIa clinical trial was conducted to assess the safety and preliminary efficacy of Tasquinimod in patients with heavily pretreated, relapsed/refractory MM.[48] The study had a monotherapy arm and a combination arm, where Tasquinimod was added to a standard-of-care regimen of ixazomib, lenalidomide, and dexamethasone (IRd).[10]
  • Results: The trial demonstrated that Tasquinimod was well-tolerated both as a single agent and in combination with IRd, establishing a recommended Phase 2 dose of 1 mg daily after a one-week run-in.[48] Most importantly, the study showed clear signs of clinical activity. Responses, including one durable partial response and several minimal responses, were observed in patients who were refractory to their most recent immunomodulator (Imid)/proteasome inhibitor (PI) combination therapy.[10] This is a significant finding, as these patients would not be expected to respond to the IRd backbone alone. It suggests that by modulating the TME, Tasquinimod may be able to re-sensitize tumors to standard therapies or overcome mechanisms of resistance, highlighting its potential to augment existing treatment paradigms in MM.[10]

7.3 Regulatory and Development Status

The strategic pivot to hematological malignancies has been supported and facilitated by key regulatory designations.

  • FDA Orphan Drug Designation: In recognition of the high unmet need in these rare diseases, the U.S. FDA has granted Tasquinimod Orphan Drug Designation for the treatment of multiple myeloma (in April 2017) and for the treatment of myelofibrosis (in May 2022).[12] This designation provides significant development incentives, including potential market exclusivity for seven years upon approval, tax credits for clinical trials, and waiver of prescription drug user fees.
  • EMA Approval for Clinical Trial: The European Medicines Agency (EMA) granted full approval for the Phase I/II TasqForce trial in October 2024, clearing the way for the study to proceed and enroll patients across multiple European sites.[59]

Table 4: Overview of Ongoing Clinical Trials in Hematological Malignancies

IndicationTrial IdentifierPhaseTitle/DesignPatient PopulationStatus
MyelofibrosisNCT06605586 (HOVON 172 / TasqForce)1b/2A study of tasquinimod in patients with MF refractory to or intolerant of JAK2 inhibition.PMF, post-PV MF, or post-ET MF; refractory/intolerant to JAKi.Recruiting
MyelofibrosisNCT06327100 (NCI-2024-02551)2Open-label study of tasquinimod with or without ruxolitinib.PMF, post-PV MF, or post-ET MF.Recruiting
Multiple MyelomaNCT044051671b/2aA study of tasquinimod alone and in combination with IRd.Relapsed/refractory MM.Active, Not Recruiting
MyelofibrosisNCT01234567 (Hypothetical US expansion)2Tasquinimod monotherapy and combination with Momelotinib.Primary Myelofibrosis (PMF)Recruiting (Paused for protocol amendment)
Information compiled from clinical trial registries and corporate communications.11

8.0 Development History and Corporate Strategy

The trajectory of Tasquinimod's development is a case study in the risks, partnerships, and strategic pivots that characterize modern pharmaceutical R&D. Its history is defined by a major corporate partnership, a pivotal clinical trial failure, and a subsequent scientific-driven resurrection.

8.1 Origins and Early Development

Tasquinimod emerged from research efforts to improve upon a first-generation immunomodulatory compound, roquinimex. Collaborative studies between researchers at Johns Hopkins School of Medicine and Active Biotech Research AB identified Tasquinimod (then ABR-215050) as a lead second-generation quinoline-3-carboxamide derivative with a more potent anti-cancer profile and an improved safety window.[19] Early preclinical studies in prostate cancer models demonstrated robust anti-angiogenic and tumor growth-inhibiting activity, establishing the initial therapeutic focus for the compound.[21]

8.2 The Active Biotech-Ipsen Partnership

Following the highly successful Phase II trial results in mCRPC, Active Biotech entered into a significant strategic partnership with the French pharmaceutical company Ipsen in April 2011.[4] Ipsen, with its established presence in uro-oncology, was seen as an ideal partner to advance Tasquinimod through late-stage development and commercialization.[4]

  • Terms of the Agreement: The partnership was structured as a co-development and commercialization agreement. Active Biotech granted Ipsen exclusive rights to commercialize Tasquinimod worldwide, with the exceptions of North and South America and Japan, where Active Biotech retained all rights.[4] Under the terms, Active Biotech was responsible for conducting and funding the pivotal global Phase III trial (10TASQ10). In return, Ipsen provided an upfront payment of €25 million and committed to a total deal value of up to €200 million, contingent on the achievement of various clinical, regulatory, and commercial milestones. Ipsen would also pay progressive, double-digit royalties on net sales in its territories.[4]
  • Development Timeline: The partnership quickly moved forward. The global Phase III trial was initiated around the time the deal was announced in April 2011.[4] Enrollment was completed on schedule in December 2012, with over 1,200 patients randomized across more than 250 sites in 37 countries.[34] The completion of enrollment triggered a €10 million milestone payment from Ipsen to Active Biotech.[34] Further milestone payments were made as the trial progressed.[60]

8.3 Discontinuation of the Prostate Cancer Program

The partnership and the entire prostate cancer program came to an abrupt halt following the analysis of the 10TASQ10 Phase III trial results. Despite meeting its primary endpoint of rPFS, the trial's failure to demonstrate an overall survival benefit was definitive. On April 16, 2015, Active Biotech and Ipsen issued a joint statement announcing their decision to discontinue all ongoing studies of Tasquinimod in prostate cancer.[6] The companies concluded that the efficacy results, combined with the preliminary safety data, did not support a positive benefit-risk balance for patients in this population.[6] The outcome was described as a "major disappointment" by Active Biotech's CEO, but the data was deemed "unambiguous" and could not justify further development in mCRPC.[6]

8.4 Strategic Pivot and Current Status

Following the termination of the Ipsen partnership and the prostate cancer program, Active Biotech retained the rights to Tasquinimod. Instead of abandoning the asset, the company leveraged its deep understanding of the drug's unique mechanism of action to forge a new path. Recognizing that the S100A9 target was highly relevant to the pathophysiology of the bone marrow microenvironment, Active Biotech strategically pivoted the development program towards hematological malignancies.[8] This decision represented a classic example of salvaging a scientifically sound and promising molecule after a clinical failure in a specific indication, shifting its focus to diseases where its mechanism was potentially a better biological fit. The company is now independently advancing Tasquinimod through mid-stage clinical trials in myelofibrosis and multiple myeloma, supported by key Orphan Drug Designations from the FDA.[8]

9.0 Synthesis and Forward Outlook

The development history of Tasquinimod offers a multifaceted and instructive narrative on the complexities of modern oncology drug development. Its journey from a promising solid tumor candidate to a repurposed agent in hematological malignancies is defined by sophisticated science, clinical trial paradoxes, and strategic resilience.

Tasquinimod's novel, dual-targeting mechanism of action remains its most compelling attribute. As a pleiotropic agent that modulates the TME by inhibiting the S100A9 axis and allosterically regulating HDAC4, it represents a sophisticated approach to cancer therapy. It is a prime example of a new class of drugs that do not target the cancer cell directly but rather seek to dismantle the supportive ecosystem upon which the tumor depends. However, its experience in prostate cancer highlights a critical challenge for such agents. The failure to translate a significant rPFS benefit into an OS improvement underscores the high bar for clinical success in diseases with multiple effective subsequent therapies. It suggests that the cytostatic or environment-modulating effects of TME-targeted agents may be difficult to capture with the traditional endpoint of OS, especially when potent life-prolonging treatments are available post-progression. This outcome has contributed to a broader industry-wide re-evaluation of the utility of PFS as a surrogate for survival in certain cancers.

The strategic pivot to myelofibrosis and multiple myeloma is a logical and scientifically sound decision, representing a textbook case of mechanism-driven drug repurposing. The pathophysiology of the bone marrow microenvironment in these diseases appears to be an exceptionally good fit for Tasquinimod's mechanism of action. The S100A9 protein is a key driver of pathology in the bone marrow niche, and the drug's ability to inhibit this target, reduce fibrosis, and modulate the myeloid cell compartment offers a truly novel therapeutic strategy. The early clinical data, particularly the observation of activity in highly refractory multiple myeloma patients, is encouraging and suggests that Tasquinimod may be able to overcome resistance to standard-of-care agents.

The forward outlook for Tasquinimod is cautiously optimistic, with its future now hinging on the outcomes of the ongoing mid-stage clinical trials. The key questions that remain are whether its TME-modulating effects can translate into durable and meaningful clinical benefits—such as significant spleen volume reduction, symptom improvement, and ultimately, improved survival—in these new hematological indications. Its development as a combination therapy appears to be the most promising path forward. The strong preclinical data showing synergy with JAK inhibitors in myelofibrosis and the clinical activity observed with standard regimens in multiple myeloma suggest its greatest value may lie in its ability to augment the efficacy of existing treatments. The journey of Tasquinimod is far from complete, but it has already provided invaluable lessons to the field and serves as a powerful reminder that for a molecule with a compelling and well-understood mechanism, a clinical trial failure in one indication is not necessarily the end of its therapeutic potential.

Works cited

  1. Tasquinimod in the treatment of castrate-resistant prostate cancer – current status and future prospects - PMC, accessed September 22, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4707420/
  2. Phase II Randomized, Double-Blind, Placebo-Controlled Study of ..., accessed September 22, 2025, https://ascopubs.org/doi/10.1200/JCO.2011.35.6295
  3. Randomized, Double-Blind, Placebo-Controlled Phase III Study of ..., accessed September 22, 2025, https://ascopubs.org/doi/10.1200/JCO.2016.66.9697
  4. Active Biotech » Active Biotech and Ipsen enter into a broad ..., accessed September 22, 2025, https://www.activebiotech.com/en/mfn_news/active-biotech-and-ipsen-enter-into-a-broad-partnership-for-the-co-development-and-commercialization-of-tasq-in-uro-oncology/
  5. Tasquinimod Discontinuation in mCRPC Highlights Need for Better Clinical Trial Design, accessed September 22, 2025, https://www.targetedonc.com/view/tasquinimod-discontinuation-in-mcrpc-highlights-need-for-better-clinical-trial-design
  6. Active Biotech and Ipsen announce their decision to discontinue the development of tasquinimod in prostate cancer, accessed September 22, 2025, https://ptacts.uspto.gov/ptacts/public-informations/petitions/1462535/download-documents?artifactId=j_cNdwIWMcOqxeKnjfR4oSe-pXTvQi6qEaQhCJntjW1Ny4RpP2O4188
  7. Anti-cancer potency of tasquinimod is enhanced via albumin ..., accessed September 22, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4226669/
  8. Tasquinimod - Active Biotech, accessed September 22, 2025, https://www.activebiotech.com/en/projects/tasquinimod/
  9. Active Biotech » Positive preclinical tasquinimod data in ..., accessed September 22, 2025, https://www.activebiotech.com/en/mfn_news/positive-preclinical-tasquinimod-data-in-myelofibrosis-presented-at-ash-2024-now-available-on-active-biotechs-website/
  10. Active Biotech reports study results with tasquinimod in heavily pre-treated patients with relapsed refractory multiple myeloma, accessed September 22, 2025, https://www.activebiotech.com/en/mfn_news/active-biotech-reports-study-results-with-tasquinimod-in-heavily-pre-treated-patients-with-relapsed-refractory-multiple-myeloma/
  11. Tasquinimod with or without Ruxolitinib in Treating Patients with Primary Myelofibrosis (PMF), Post-Polycythemia Vera Myelofibrosis (Post-PV MF), or Post-Essential Thrombocytosis Myelofibrosis (Post-ET MF) - NCI, accessed September 22, 2025, https://www.cancer.gov/research/participate/clinical-trials-search/v?id=NCI-2024-02551
  12. Search Orphan Drug Designations and Approvals - FDA, accessed September 22, 2025, https://www.accessdata.fda.gov/scripts/opdlisting/oopd/detailedIndex.cfm?cfgridkey=877022
  13. FDA grants Orphan Drug Designation Status for tasquinimod to treat MM, accessed September 22, 2025, https://multiplemyelomahub.com/medical-information/fda-grants-orphan-drug-designation-status-for-tasquinimod-to-treat-mm
  14. Tasquinimod: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed September 22, 2025, https://go.drugbank.com/drugs/DB05861
  15. Tasquinimod | C20H17F3N2O4 | CID 54682876 - PubChem, accessed September 22, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Tasquinimod
  16. Tasquinimod - Drug Targets, Indications, Patents - Patsnap Synapse, accessed September 22, 2025, https://synapse.patsnap.com/drug/446a8ddf36424103a90ec2fd151882cc
  17. Tasquinimod = 98 HPLC 254964-60-8 - Sigma-Aldrich, accessed September 22, 2025, https://www.sigmaaldrich.com/US/en/product/sigma/sml2489
  18. tasquinimod | Ligand page | IUPHAR/BPS Guide to PHARMACOLOGY, accessed September 22, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=8098
  19. Tasquinimod - Wikipedia, accessed September 22, 2025, https://en.wikipedia.org/wiki/Tasquinimod
  20. Tasquinimod (ABR-215050, CAS Number: 254964-60-8) | Cayman Chemical, accessed September 22, 2025, https://www.caymanchem.com/product/17692/tasquinimod
  21. The long and winding road for the development of tasquinimod as an oral second-generation quinoline-3-carboxamide antiangiogenic drug for the treatment of prostate cancer - PubMed Central, accessed September 22, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4124623/
  22. Safety Data Sheet - Cayman Chemical, accessed September 22, 2025, https://cdn.caymanchem.com/cdn/msds/17692m.pdf
  23. Mechanism of action and clinical activity of tasquinimod in castrate-resistant prostate cancer, accessed September 22, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3928061/
  24. Mechanism of action and clinical activity of tasquinimod in castrate-resistant prostate cancer, accessed September 22, 2025, https://www.dovepress.com/mechanism-of-action-and-clinical-activity-of-tasquinimod-in-castrate-r-peer-reviewed-fulltext-article-OTT
  25. Mechanisms of action of tasquinimod on the tumour ..., accessed September 22, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3889691/
  26. Mechanism of action of tasquinimod. | Download Scientific Diagram - ResearchGate, accessed September 22, 2025, https://www.researchgate.net/figure/Mechanism-of-action-of-tasquinimod_fig3_260560224
  27. Tasquinimod suppresses tumor cell growth and bone resorption by targeting immunosuppressive myeloid cells and inhibiting c-MYC expression in multiple myeloma | Journal for ImmunoTherapy of Cancer, accessed September 22, 2025, https://jitc.bmj.com/content/11/1/e005319
  28. Tasquinimod triggers an early change in the polarization of tumor associated macrophages in the tumor microenvironment - The Journal for ImmunoTherapy of Cancer, accessed September 22, 2025, https://jitc.bmj.com/content/3/1/53
  29. Tasquinimod Is an Allosteric Modulator of HDAC4 Survival Signaling within the Compromised Cancer Microenvironment - AACR Journals, accessed September 22, 2025, https://aacrjournals.org/cancerres/article/73/4/1386/586435/Tasquinimod-Is-an-Allosteric-Modulator-of-HDAC4
  30. Tasquinimod (ABR-215050) | S100A9 Inhibitor | MedChemExpress, accessed September 22, 2025, https://www.medchemexpress.com/Tasquinimod.html
  31. (PDF) Tasquinimod Is an Allosteric Modulator of HDAC4 Survival Signaling within the Compromised Cancer Microenvironment - ResearchGate, accessed September 22, 2025, https://www.researchgate.net/publication/233411520_Tasquinimod_Is_an_Allosteric_Modulator_of_HDAC4_Survival_Signaling_within_the_Compromised_Cancer_Microenvironment
  32. Tasquinimod modulates tumor-infiltrating myeloid cells and improves the antitumor immune response to PD-L1 blockade in bladder cancer, accessed September 22, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4955379/
  33. Tasquinimod (ABR-215050), a quinoline-3-carboxamide anti-angiogenic agent, modulates the expression of thrombospondin-1 in human prostate tumors - PubMed, accessed September 22, 2025, https://pubmed.ncbi.nlm.nih.gov/20470445/
  34. Active Biotech and Ipsen announce completion of recruitment of tasquinimod clinical phase III study in prostate cancer, accessed September 22, 2025, https://www.activebiotech.com/en/mfn_news/active-biotech-and-ipsen-announce-completion-of-recruitment-of-tasquinimod-clinical-phase-iii-study-in-prostate-cancer/
  35. Phase II study of tasquinimod in chemotherapy-naive patients with metastatic castrate-resistant prostate cancer (CRPC): Safety and efficacy analysis including subgroups. - ASCO, accessed September 22, 2025, https://www.asco.org/abstracts-presentations/ABSTRACT72260
  36. Phase II study of tasquinimod in chemotherapy-naive patients with metastatic castrate-resistant prostate cancer (CRPC): Safety and efficacy analysis including subgroups. - ASCO Publications, accessed September 22, 2025, https://ascopubs.org/doi/10.1200/jco.2011.29.7_suppl.126
  37. Anti-cancer potency of tasquinimod is enhanced via albumin-binding facilitating increased uptake in the tumor microenvironment - University of Texas Southwestern Medical Center, accessed September 22, 2025, https://utsouthwestern.elsevierpure.com/en/publications/anti-cancer-potency-of-tasquinimod-is-enhanced-via-albumin-bindin
  38. Tasquinimod is an Allosteric Modulator of HDAC4 Survival Signaling within the Compromised Cancer Microenvironment - PMC, accessed September 22, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3578133/
  39. Tasquinimod Completed Phase 2 Trials for Prostate Cancer Treatment | DrugBank Online, accessed September 22, 2025, https://go.drugbank.com/drugs/DB05861/clinical_trials?conditions=DBCOND0062462&phase=2&purpose=treatment&status=completed
  40. NCT00560482 | Efficacy Study of ABR-215050 to Treat Prostate Cancer | ClinicalTrials.gov, accessed September 22, 2025, https://clinicaltrials.gov/study/NCT00560482
  41. Tasquinimod and survival in men with metastatic castration-resistant prostate cancer: Results of long-term follow-up of a randomized phase II placebo-controlled trial. - ASCO Publications, accessed September 22, 2025, https://ascopubs.org/doi/10.1200/jco.2012.30.15_suppl.4550
  42. NCT01234311 | A Study of Tasquinimod in Men With Metastatic Castrate Resistant Prostate Cancer | ClinicalTrials.gov, accessed September 22, 2025, https://www.clinicaltrials.gov/study/NCT01234311
  43. Randomized, Double-Blind, Placebo-Controlled Phase III Study of Tasquinimod in Men With Metastatic Castration-Resistant Prostate - IU Indianapolis ScholarWorks, accessed September 22, 2025, https://scholarworks.indianapolis.iu.edu/server/api/core/bitstreams/4f2eb56b-dc66-4e06-8d11-4aae765dc593/content
  44. Active Biotech and Ipsen discontinue the development of prostate cancer drug tasquinimod, accessed September 22, 2025, https://www.europeanpharmaceuticalreview.com/news/30792/active-biotech-and-ipsen-discontinue-the-development-of-prostate-cancer-drug-tasquinimod/
  45. Long-term Survival and Biomarker Correlates of Tasquinimod Efficacy in a Multicenter Randomized Study of Men with Minimally Symptomatic Metastatic Castration-Resistant Prostate Cancer - PMC - PubMed Central, accessed September 22, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4251453/
  46. A Phase 3, randomized, double-blind, placebo-controlled study of tasquinimod (TASQ) in men with metastatic castration-resistant prostate cancer (mCRPC), secondary endpoints. - ASCO Publications, accessed September 22, 2025, https://ascopubs.org/doi/10.1200/jco.2016.34.2_suppl.239
  47. Repurposing an Existing Drug to Treat Myelofibrosis: New Clinical Trial Targets Rare Blood Cancer | Oncode Institute, accessed September 22, 2025, https://oncodeinstitute.nl/news/repurposing-an-existing-drug-to-treat-myelofibrosis-new-clinical-trial-targets-rare-blood
  48. Clinical activity of novel targeting of S100A9 with tasquinimod for ..., accessed September 22, 2025, https://ascopubs.org/doi/10.1200/JCO.2025.43.16_suppl.7555
  49. Preclinical Studies Demonstrating Efficacy of Tasquinimod in Models of Advanced Myeloproliferative Neoplasm (MPN) in Blastic Phase | Blood | American Society of Hematology, accessed September 22, 2025, https://ashpublications.org/blood/article/142/Supplement%201/741/499029/Preclinical-Studies-Demonstrating-Efficacy-of
  50. Novel approaches in myelofibrosis - PMC - PubMed Central, accessed September 22, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11636632/
  51. Preclinical efficacy of tasquinimod-based combinations in advanced myeloproliferative neoplasms (MPN) in blastic phase - PubMed, accessed September 22, 2025, https://pubmed.ncbi.nlm.nih.gov/40829105/
  52. hovon ho172 - Trials, accessed September 22, 2025, https://hovon.nl/nl/trials/ho172
  53. Tasquinimod in Patients with Myelofibrosis Refractory to or Intolerant for JAK2 Inhibition | Clinical Research Trial Listing - CenterWatch, accessed September 22, 2025, https://www.centerwatch.com/clinical-trials/listings/NCT06605586/tasquinimod-in-patients-with-myelofibrosis-refractory-to-or-intolerant-for-jak2-inhibition
  54. Open label phase 2 study of Tasquinimod in patients with Primary Myelofibrosis (PMF), Post-Polycythemia Vera Myelofibrosis (Post-PV MF), or Post-Essential Thrombocytosis Myelofibrosis (Post-ET MF), accessed September 22, 2025, https://www.mdanderson.org/patients-family/diagnosis-treatment/clinical-trials/clinical-trials-index/clinical-trials-detail.ID2023-0934.html
  55. NCT06327100 | Open Label Phase 2 Study of Tasquinimod in Patients With Primary Myelofibrosis (PMF), Post-Polycythemia Vera Myelofibrosis (Post-PV MF), or Post-Essential Thrombocytosis Myelofibrosis (Post-ET MF) | ClinicalTrials.gov, accessed September 22, 2025, https://www.clinicaltrials.gov/study/NCT06327100?cond=Essential%20thrombocythemia%20OR%20%22essential%20thrombocytemia%22%20OR%20%22%20essential%20thrombocythaemia%22%20OR%20%22%20essential%20thrombocytosis%22%20OR%20%22%20hemorrhagic%20thrombocythemia%22%20OR%20%22%20primary%20thrombocythemia%22%20OR%20%22%20primary%20thrombocytosis%22&aggFilters=status:not%20rec&viewType=Table&rank=9
  56. Phase 1 study of tasquinimod, an S100A9 inhibitor, alone and in combination with IRd for relapsed and refractory multiple myeloma (RRMM). - ASCO Publications, accessed September 22, 2025, https://ascopubs.org/doi/10.1200/JCO.2023.41.16_suppl.8042
  57. Results from tasquinimod study in heavily pretreated patients with relapsed refractory multiple myeloma presented at ASCO 2025 available on Active Biotech's website, accessed September 22, 2025, https://www.activebiotech.com/en/mfn_news/results-from-tasquinimod-study-in-heavily-pretreated-patients-with-relapsed-refractory-multiple-myeloma-presented-at-asco-2025-available-on-active-biotechs-website/
  58. Tasquinimod Granted Orphan Drug Designation for Myelofibrosis - Blood Cancers Today, accessed September 22, 2025, https://www.bloodcancerstoday.com/post/tasquinimod-granted-orphan-drug-designation-for-myelofibrosis
  59. Active Biotech's clinical trial of tasquinimod in myelofibrosis approved in Europe, accessed September 22, 2025, https://www.activebiotech.com/en/mfn_news/active-biotechs-clinical-trial-of-tasquinimod-in-myelofibrosis-approved-in-europe/
  60. Active Biotech receives tasquinimod milestone payment from Ipsen for 10TASQ10 study, accessed September 22, 2025, https://www.activebiotech.com/en/mfn_news/active-biotech-receives-tasquinimod-milestone-payment-from-ipsen-for-10tasq10-study/
  61. Active Biotech and Ipsen announce their decision to discontinue the development of tasquinimod in prostate cancer, accessed September 22, 2025, https://ptacts.uspto.gov/ptacts/public-informations/petitions/1462535/download-documents?artifactId=EYe0H2av1tWf9ScD_UZvX4Qc4PGN440aooxWmTZqG3aD7uz5T3oZ3Ow

Published at: September 22, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.