MedPath

Tinostamustine Advanced Drug Monograph

Published:Oct 20, 2025

Generic Name

Tinostamustine

Drug Type

Small Molecule

Chemical Formula

C19H28Cl2N4O2

CAS Number

1236199-60-2

Comprehensive Report on the Investigational Antineoplastic Agent: Tinostamustine (EDO-S101)

Executive Summary

Tinostamustine, also known by its code designation EDO-S101, is a first-in-class, investigational small molecule antineoplastic agent engineered with a unique bifunctional mechanism of action.[1] It is a novel chemical entity created by fusing the cytotoxic alkylating agent bendamustine with the pan-histone deacetylase inhibitor (HDACi) vorinostat.[1] This rational design is intended to overcome common mechanisms of cancer resistance by simultaneously inducing direct DNA damage while epigenetically modulating the tumor microenvironment to enhance drug accessibility and cellular vulnerability.

Preclinical validation has demonstrated potent antitumor activity across a broad spectrum of hematologic malignancies and solid tumors. In vitro, Tinostamustine inhibits Class I and II histone deacetylases at nanomolar concentrations, induces apoptosis in cancer cell lines irrespective of p53 status or prior resistance to other alkylating agents, and shows superior cytotoxicity compared to its parent compounds administered individually.[3] A key preclinical feature is its excellent penetration of the central nervous system (CNS), providing a strong rationale for its investigation in brain malignancies.[7] Furthermore, its mechanism extends to immunomodulation, upregulating targets like CD38 on myeloma cells, thereby creating a synergistic potential with antibody-based immunotherapies such as daratumumab.[9]

The clinical development program, which has advanced to Phase II, has investigated Tinostamustine as a monotherapy in heavily pre-treated patient populations with high unmet medical needs. In relapsed/refractory hematologic malignancies (NCT02576496), the agent has demonstrated clinically meaningful signals of efficacy, with an overall response rate (ORR) of 37% in Hodgkin's lymphoma and 50% in cutaneous T-cell lymphoma.[11] In advanced solid tumors (NCT03345485), while objective responses are more modest, the achievement of durable stable disease in a significant portion of patients for whom no other standard therapies are available underscores its clinical benefit.[15]

The safety profile of Tinostamustine is consistent and has been characterized as predictable and manageable.[11] The most common and clinically significant treatment-emergent adverse events are hematological toxicities, particularly thrombocytopenia, and QTc interval prolongation, both of which are monitorable and can be managed with dose adjustments and supportive care.[11]

Strategically, Tinostamustine is being positioned to address critical unmet needs in oncology. It has received Orphan Drug Designation from both the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA) for the treatment of T-cell prolymphocytic leukemia (T-PLL), a rare and aggressive malignancy with few effective options.[18] Its inclusion in the innovative GBM AGILE adaptive trial for glioblastoma, particularly for the difficult-to-treat MGMT-unmethylated subtype, highlights its potential in CNS oncology.[21] Supported by a robust intellectual property portfolio, Tinostamustine represents a promising therapeutic candidate whose true potential may be realized both as a monotherapy in niche indications and as a powerful combination partner capable of sensitizing tumors to other anticancer agents.

Drug Profile and Mechanism of Action

Chemical Identity and Properties

Tinostamustine is a synthetic small molecule classified as an antineoplastic agent.[22] Its development represents a targeted effort to create a multi-action therapy within a single chemical structure. The fundamental chemical and physical properties of Tinostamustine are critical for understanding its pharmacological behavior and are summarized below.

The generic name for the compound is Tinostamustine, which has been adopted by the USAN council.[24] Throughout its development, it has been referred to by several code designations and synonyms, most prominently EDO-S101, EDO-S 101, EDO-S-101, and NL-101.[2] It is also described functionally as a bendamustine-vorinostat fusion molecule.[2]

The formal chemical name under IUPAC nomenclature is 7-[bis(2-chloroethyl)amino]-1-methylbenzimidazol-2-yl]-N-hydroxyheptanamide.[1] Its unique structure is registered under the CAS Number 1236199-60-2.[1] The molecular formula of Tinostamustine is $C_{19}H_{28}Cl_{2}N_{4}O_{2}$, corresponding to a molecular weight of approximately 415.4 g/mol.[1] These core identifiers are essential for its unambiguous identification in scientific literature, regulatory filings, and chemical databases.

Table 1: Key Drug Identifiers and Physicochemical Properties

Identifier/PropertyValueSource(s)
DrugBank IDDB151471
TypeSmall Molecule1
CAS Number1236199-60-21
IUPAC Name7-[bis(2-chloroethyl)amino]-1-methylbenzimidazol-2-yl]-N-hydroxyheptanamide1
Molecular Formula$C_{19}H_{28}Cl_{2}N_{4}O_{2}$1
Molecular Weight415.4 g/mol1
UNII29DKI2H2NY1
SMILESCN1C2=C(C=C(C=C2)N(CCCl)CCCl)N=C1CCCCCCC(=O)NO1
InChIKeyGISXTRIGVCKQBX-UHFFFAOYSA-N1

A Novel Dual-Action Therapeutic Strategy

Tinostamustine was conceived as a first-in-class alkylating deacetylase inhibitor (AK-DACi), representing a paradigm of rational drug design that integrates two distinct and potentially synergistic antineoplastic mechanisms into a single new chemical entity.[1] The molecule is a covalent fusion of two well-characterized parent compounds: the bifunctional alkylating agent bendamustine and the pan-histone deacetylase inhibitor (HDACi) vorinostat (also known as SAHA).[1]

The central hypothesis underpinning this design is to overcome intrinsic and acquired resistance to conventional chemotherapy by creating a more favorable environment for DNA damage.[28] In many cancer cells, DNA is tightly wound around histone proteins in a condensed chromatin structure (heterochromatin), which can physically shield it from the damaging effects of alkylating agents. The vorinostat component of Tinostamustine is designed to first address this barrier by inhibiting histone deacetylases, leading to a more relaxed, open chromatin state (euchromatin). This epigenetic "priming" is intended to improve the access of the bendamustine component to the DNA strands, thereby potentiating its ability to induce cytotoxic DNA damage and cell death.[15] This integrated, sequential mechanism within a single molecule aims to deliver a superior therapeutic effect compared to the administration of either agent alone or as a simple non-covalent combination.

The Bendamustine Moiety: DNA Damage and Cytotoxicity

The cytotoxic activity of Tinostamustine is driven in part by its bendamustine moiety, which functions as a classical DNA alkylating agent.[1] This component contains a nitrogen mustard group, which is highly reactive and capable of forming covalent bonds with nucleophilic sites on various macromolecules, most critically DNA and RNA.[1]

Upon administration, the bendamustine portion of the molecule alkylates DNA, leading to the formation of adducts. Because it is a bifunctional alkylator, it can form crosslinks both within a single DNA strand (intra-strand) and between the two strands of the DNA helix (inter-strand), as well as crosslinks between DNA and proteins.[3] These lesions are highly cytotoxic. They physically obstruct the DNA replication and transcription machinery, effectively halting the synthesis of new DNA, RNA, and proteins essential for cell survival and proliferation.[1] The accumulation of this extensive and irreparable DNA damage serves as a powerful signal to initiate programmed cell death, ultimately resulting in tumor cell apoptosis.[1]

The Vorinostat Moiety: Epigenetic Modulation and Apoptosis

The second functional component of Tinostamustine is its vorinostat moiety, which provides a potent epigenetic mechanism of action.[3] Vorinostat is a well-established pan-HDAC inhibitor, meaning it broadly targets multiple isoforms of histone deacetylase enzymes, specifically those in Class I (HDAC1, 2, 3, 8) and Class II (HDAC6, 10).[3]

Histone deacetylases play a crucial role in gene regulation by removing acetyl groups from lysine residues on the tails of histone proteins. This deacetylation increases the positive charge of the histones, strengthening their interaction with the negatively charged phosphate backbone of DNA and leading to a condensed, transcriptionally silent chromatin structure.[33] HDACs are often overexpressed in cancer cells, contributing to the silencing of tumor suppressor genes.

By inhibiting these enzymes, the vorinostat moiety of Tinostamustine blocks this deacetylation process. The result is an accumulation of acetylated histones, a state known as hyperacetylation.[1] This neutralizes the positive charge on the histone tails, weakening their grip on DNA and causing the chromatin to relax into a more open and transcriptionally active conformation.[33] This "chromatin remodeling" has profound downstream effects: it can reactivate the expression of silenced tumor suppressor genes (e.g., p21), which in turn can induce cell cycle arrest, inhibit tumor cell division, and trigger apoptosis.[1] Thus, the vorinostat moiety contributes to the drug's antineoplastic activity through an independent, epigenetically driven apoptotic pathway.

Synergistic Antineoplastic Effects and UPR Induction

The therapeutic innovation of Tinostamustine lies not in the individual actions of its components, but in their synergistic interplay, which is designed to overcome key mechanisms of therapeutic resistance. The dual-action mechanism is not merely additive; it is a carefully orchestrated sequence where one action potentiates the other. The primary synergistic hypothesis is that the epigenetic modulation by the vorinostat moiety directly enhances the cytotoxic efficacy of the bendamustine moiety. Cancer cells often protect their DNA from alkylating agents by maintaining a tightly packed chromatin state. The vorinostat component acts first, inhibiting HDACs to induce histone hyperacetylation and chromatin relaxation.[28] This decondensation effectively "unmasks" the DNA, making it significantly more accessible to the alkylating nitrogen mustard group of the bendamustine component.[15] This improved access allows for more efficient and extensive DNA alkylation and cross-linking, leading to a level of DNA damage and subsequent apoptosis that is superior to what could be achieved by either agent alone or their co-administration as separate drugs.[1] This synergistic mechanism provides a compelling explanation for the preclinical findings that Tinostamustine retains activity in cell lines resistant to other chemotherapies, including bendamustine itself, and is effective in glioblastoma models regardless of MGMT expression, a common resistance pathway for traditional alkylators.[5]

Beyond this primary synergy, Tinostamustine induces a distinct cellular vulnerability through its specific inhibition of HDAC6. This action has been shown to trigger the activation of inositol-requiring enzyme 1 (IRE-1), a key sensor protein in the endoplasmic reticulum that governs the unfolded protein response (UPR).[1] The UPR is a cellular stress response pathway activated by an accumulation of misfolded proteins. While it initially aims to restore cellular homeostasis, chronic or overwhelming UPR activation can pivot to induce apoptosis. Critically, the induction of the UPR by Tinostamustine has been found to sensitize cancer cells to other classes of cytotoxic agents, most notably proteasome inhibitors like bortezomib and carfilzomib.[1] Proteasome inhibitors function by blocking the cell's primary machinery for degrading misfolded proteins, which itself induces significant proteotoxic stress. By first activating the UPR with Tinostamustine, the cell's stress pathways are already primed, making it exquisitely sensitive to the subsequent blockade of protein degradation by a proteasome inhibitor. This provides a strong mechanistic basis for the significant synergistic cytotoxicity observed in preclinical studies combining Tinostamustine with proteasome inhibitors in multiple myeloma cells.[4]

Preclinical Development and Scientific Validation

In Vitro and Ex Vivo Antitumor Activity

The biological activity of Tinostamustine was extensively characterized in preclinical laboratory models prior to human studies. These experiments established its potency as a dual-action agent and confirmed its cytotoxic effects across a range of cancer types.

As an HDAC inhibitor, Tinostamustine demonstrates potent, broad-spectrum activity. Cell-free assays have quantified its inhibitory concentration ($IC_{50}$) against multiple HDAC isoforms, confirming its designation as a pan-HDACi. It potently inhibits Class I enzymes, including HDAC1 ($IC_{50}$ = 9 nM), HDAC2 ($IC_{50}$ = 9 nM), HDAC3 ($IC_{50}$ = 25 nM), and HDAC8 ($IC_{50}$ = 107 nM). It is also highly active against Class II enzymes, particularly HDAC6 ($IC_{50}$ = 6 nM) and HDAC10 ($IC_{50}$ = 72 nM).[3] This potent, low-nanomolar inhibition across multiple key HDACs underpins its ability to induce widespread epigenetic changes in cancer cells.

Table 2: Summary of HDAC Inhibition Profile ($IC_{50}$ Values)

HDAC IsoformClassIC50​ (nM)Source(s)
HDAC1I94
HDAC2I94
HDAC3I254
HDAC6II64
HDAC8I1074
HDAC10II724

In cell-based assays, Tinostamustine has shown robust antitumor activity in models of hematologic malignancies. It effectively reduces the growth of multiple myeloma (MM) cell lines, with $IC_{50}$ values ranging from 1.6 to 4.8 µM. Notably, this activity was observed regardless of the cells' p53 mutational status or pre-existing resistance to other DNA alkylating agents, highlighting its potential to overcome common resistance mechanisms.[3] Its efficacy was further confirmed in ex vivo studies using bone marrow samples from patients with both early- and late-stage MM, where it successfully induced cancer cell death.[3] Strong antitumor activity, characterized by the induction of apoptosis via cleavage of caspases 3 and 9, has also been demonstrated in other hematologic cancer cell lines, including HL-60 (acute promyelocytic leukemia) and Daudi (Burkitt's lymphoma).[32]

The compound's activity extends to solid tumors. In preclinical models of glioblastoma (GBM), Tinostamustine exhibited dose-dependent cytotoxicity against both temozolomide-sensitive (U-87 MG) and temozolomide-resistant (U-138 MG) cell lines, with significant cell killing observed at a concentration of 5 µM.[28] Broad activity has also been documented in preclinical models of sarcoma, small-cell lung cancer (SCLC), breast cancer, and ovarian cancer.[35] A consistent finding across these studies is the superior efficacy of the fusion molecule. In GBM models, Tinostamustine demonstrated stronger antiproliferative and pro-apoptotic effects than either vorinostat or bendamustine administered alone, with an efficacy comparable to their combination, underscoring the success of its bifunctional design.[5]

In Vivo Efficacy in Animal Models

The promising in vitro activity of Tinostamustine was subsequently validated in in vivo animal models, demonstrating its therapeutic potential in a whole-organism context. These studies provided crucial evidence of its ability to control tumor growth and improve survival.

In models of multiple myeloma, Tinostamustine showed significant efficacy. In a mouse xenograft model using MM1S cells, weekly administration of Tinostamustine at 60 mg/kg effectively reduced tumor growth and prolonged the survival of the animals.[3] Its potential in difficult-to-treat disease was further substantiated in a multidrug-resistant Vk12653 mouse transplant model of refractory MM, where it also conferred a significant survival benefit.[3]

The compound's efficacy against brain tumors was tested in highly relevant orthotopic models of glioblastoma, where tumor cells are implanted directly into the brain. In these challenging models, Tinostamustine monotherapy significantly suppressed tumor growth and prolonged both disease-free survival (DFS) and overall survival (OS).[5] Its performance was notably superior to that of its parent compound bendamustine, as well as to standard treatments like radiotherapy (RT) and temozolomide, establishing its potent anti-GBM activity in vivo.[5]

In models of lymphoma, the antitumor effects were equally striking. Following intravenous administration to mice bearing subcutaneous human Burkitt's lymphoma tumors, the tumors were observed to rapidly shrink or, in some cases, be completely eradicated.[32] These in vivo results across multiple cancer types provided a strong foundation for advancing Tinostamustine into clinical trials in humans.

Preclinical Pharmacokinetics and CNS Distribution

Preclinical studies of Tinostamustine's absorption, distribution, metabolism, and excretion (ADME) profile have revealed key characteristics that influence its therapeutic application, most notably its distribution to the central nervous system (CNS).

A critical and differentiating feature of Tinostamustine is its ability to effectively cross the blood-brain barrier (BBB), a major obstacle for many systemic cancer therapies. Preclinical pharmacokinetic studies have demonstrated excellent CNS penetration. Following intravenous administration, the brain-to-plasma concentration ratio was found to be 13.8% for infusion and 16.5% for bolus administration.[7] This substantial distribution into the CNS is a crucial attribute, as it allows the drug to reach therapeutic concentrations within the brain. This property provides a direct and compelling scientific rationale for its clinical investigation in primary brain tumors like glioblastoma, where the BBB often renders other systemic agents ineffective.[21] The observed superiority of Tinostamustine over temozolomide in preclinical orthotopic brain tumor models can be partly attributed to this efficient CNS penetration.[5]

Regarding its metabolism and elimination, Tinostamustine exhibits a short plasma half-life. In studies conducted in beagle dogs, the half-life was approximately 5-10 minutes, with plasma concentrations declining rapidly after the cessation of intravenous infusion.[38] In vitro metabolic studies using cryopreserved hepatocytes from humans, rats, and dogs indicate that the primary metabolic pathways for Tinostamustine involve oxidation of the hydroxy amide moiety to a carboxylic acid and hydrolysis of one of the chloroethyl side chains.[38]

Immunomodulatory Potential

In addition to its direct cytotoxic and epigenetic effects on tumor cells, preclinical research has uncovered a significant immunomodulatory role for Tinostamustine, suggesting its potential as a powerful agent for combination with immunotherapy. This activity stems from its ability to alter the cell surface protein expression of cancer cells, making them more recognizable and susceptible to immune-mediated killing.

This effect has been most clearly demonstrated in the context of multiple myeloma. Studies have shown that treatment with Tinostamustine leads to a significant increase in the expression of CD38 on the surface of myeloma cell lines.[9] This is particularly relevant because CD38 is the target of the highly effective monoclonal antibody therapy, daratumumab. The mechanism for this upregulation is directly linked to the drug's HDACi function; the increased expression of CD38 occurs in parallel with an increase in histone H3 acetylation at the CD38 gene locus, indicating that Tinostamustine epigenetically reactivates or enhances the transcription of the CD38 gene.[9]

By increasing the density of the target antigen on the tumor cell surface, Tinostamustine effectively "primes" the cancer cells for attack by daratumumab. Preclinical experiments have confirmed this functional consequence: pretreatment of myeloma cells with Tinostamustine enhanced their sensitivity to daratumumab-mediated cytotoxicity in vitro.[9] Furthermore, Tinostamustine also augments the expression of MICA and MICB, which are ligands for the activating receptor NKG2D found on natural killer (NK) cells and other immune effector cells.[10] This further increases the immunogenicity of the tumor cells. The combination of Tinostamustine followed by daratumumab administration in in vivo mouse models resulted in significantly delayed tumor growth and improved survival compared to either agent alone.[9] This ability to epigenetically reprogram the tumor cell surface to enhance vulnerability to antibody-based and cell-mediated immune attack provides a strong rationale for exploring Tinostamustine as a combination partner for a wide range of immunotherapies.

Clinical Development Program and Efficacy

Overview of Clinical Trials

The clinical development of Tinostamustine has progressed through Phase I and II studies, focusing on patient populations with advanced hematologic malignancies and solid tumors for whom standard therapeutic options are limited or exhausted. The program has been designed to first establish the safety, tolerability, and recommended dose of the agent, and then to explore its efficacy signals across a range of cancer types. The maximum clinical trial phase completed or ongoing is Phase II.[1] The major clinical trials forming the basis of the current understanding of Tinostamustine are summarized in Table 3.

Table 3: Overview of Major Clinical Trials for Tinostamustine

NCT NumberOfficial TitlePhaseCondition(s)SponsorStatus (as of late 2024)Source(s)
NCT02576496A Phase 1 Trial to Investigate the Safety, Pharmacokinetic Profiles and the Efficacy of Tinostamustine...in Relapsed/Refractory Hematologic MalignanciesPhase 1Hematologic Malignancies (MM, HL, PTCL, CTCL, T-PLL)Mundipharma Research LimitedActive, not recruiting7
NCT03345485A Phase 1/2 Study to Investigate the Safety, Pharmacokinetics and Efficacy of EDO-S101...in Patients With Advanced Solid TumorsPhase 1/2Solid Tumors (SCLC, Sarcoma, TNBC, Ovarian, Endometrial)Mundipharma Research LimitedActive, not recruiting7
N/A (GBM AGILE)Glioblastoma Adaptive Global Innovative Learning EnvironmentPhase 2/3Glioblastoma (Newly Diagnosed and Recurrent)Global Coalition for Adaptive ResearchRecruiting (Tinostamustine arm pending activation)21
NCI-2018-00872Tinostamustine with or without Radiation Therapy in Treating Patients with Newly Diagnosed MGMT-Unmethylated GlioblastomaPhase 1MGMT-Unmethylated GlioblastomaNational Cancer Institute (NCI)Recruiting37

Efficacy in Relapsed/Refractory Hematologic Malignancies (NCT02576496)

The first-in-human study of Tinostamustine (NCT02576496) was a multi-center, open-label, Phase 1 trial conducted in patients with relapsed/refractory (R/R) hematologic malignancies.[11] The study was structured in two stages: a dose-escalation stage (Stage 1) to determine the safety profile and maximum tolerated dose (MTD), followed by a cohort-expansion stage (Stage 2) to further evaluate safety and preliminary efficacy at the recommended Phase 2 dose (RP2D).

In Stage 1, doses were escalated in a standard 3+3 design, starting from a 1-hour infusion of 20 mg/m² up to a maximum dose of 150 mg/m².[40] Based on the safety data from this stage, the MTD was established as 100 mg/m² administered as a 60-minute intravenous infusion.[11]

Stage 2 enrolled specific cohorts of patients to receive the RP2D. The dosing schedule varied by disease: patients with lymphoma received treatment on Day 1 of a 21-day cycle, while multiple myeloma patients were treated on Days 1 and 15 of a 28-day cycle.[40] The efficacy results from the expansion cohorts in this heavily pre-treated population were promising:

  • Hodgkin's Lymphoma (HL): In a cohort of 20 patients with R/R HL who had received a median of 4.5 prior lines of therapy, Tinostamustine demonstrated significant clinical activity. The Overall Response Rate (ORR) was 37%, which included 2 patients (10%) achieving a Complete Response (CR) and 5 patients (25%) achieving a Partial Response (PR). The median Progression-Free Survival (PFS) in this cohort was 3.8 months (95% CI: 2.2–9.4 months).[11]
  • Cutaneous T-Cell Lymphoma (CTCL): A cohort of 13 patients (12 evaluable for efficacy) with advanced mycosis fungoides or Sézary syndrome showed a strong response to treatment. The ORR was 50% (6 of 12 patients), comprising 2 CRs and 4 PRs. The Clinical Benefit Rate (CBR), which includes patients with stable disease for at least 4 cycles, was 83.3%. The median PFS in the CTCL cohort was 7.3 months (95% CI: 3.2 months–Not Estimable).[12]
  • Overall Efficacy (All Cohorts): Across 46 evaluable patients in the expansion phase (including cohorts for HL, CTCL, peripheral T-cell lymphoma, T-cell prolymphocytic leukemia, and multiple myeloma [MM]), the aggregate ORR was 30.4% (95% CI: 17.7%–45.8%) and the CBR was 52.2% (95% CI: 36.9%–67.1%). The overall median PFS for this diverse population was 4.3 months (95% CI: 2.7–6.6 months).[44]

These results indicate that Tinostamustine monotherapy has clear biological activity and provides clinical benefit in a difficult-to-treat population of patients with various R/R hematologic cancers.

Efficacy in Advanced Solid Tumors (NCT03345485)

Building on the preclinical data in solid tumors, a Phase 1/2 study (NCT03345485) was initiated to evaluate Tinostamustine in patients with advanced or metastatic solid tumors who had progressed on prior therapies and had no other available standard treatment options.[7] The study followed a similar two-part design with a dose-escalation phase followed by cohort expansions.

The Phase 1 dose-escalation portion enrolled 22 patients across six ascending dose cohorts, from 60 mg/m² to 100 mg/m², administered on Days 1 and 15 of a 28-day cycle.[17] Preliminary efficacy at the 16-week follow-up showed a partial response in 4.5% of patients (1 patient) and stable disease in 36.4% of patients.[17] Based on the safety and pharmacokinetic data, the Recommended Phase 2 Dose (RP2D) was determined to be 80 mg/m² administered as a 1-hour infusion on Days 1 and 15 of each 4-week cycle.[15]

The Phase 2 portion evaluated this RP2D in expansion cohorts for five specific tumor types: small-cell lung cancer (SCLC), soft tissue sarcoma (STS), triple-negative breast cancer (TNBC), ovarian cancer, and endometrial cancer. In 36 patients evaluable for response, the efficacy signals, while modest, were clinically relevant given the advanced nature of the disease in this population.

  • Two patients (5.6%) achieved a partial response: one with synovial sarcoma and one with ovarian cancer.[15]
  • A total of 14 patients (38.9%) achieved stable disease for a duration of at least 4 months.[15]
  • The overall Clinical Benefit Rate (ORR + SD ≥4 months) was 44.4%.[15]
  • For all treated patients, the median PFS was 2.2 months (95% CI: 1.8–3.3 months) and the median overall survival (OS) was 5.5 months (95% CI: 4.1–15.0 months).[15]

These outcomes must be interpreted within the context of the study's inclusion criteria, which enrolled patients with very limited or no other therapeutic options.[15] In such a heavily pre-treated, end-stage population, achieving durable disease stabilization for over four months in nearly 40% of patients represents a meaningful clinical benefit. The results, therefore, support the biological activity of Tinostamustine in solid tumors and warrant further investigation, potentially in earlier lines of therapy or in rational combination regimens where its unique mechanism can be further exploited.

Investigation in Glioblastoma (GBM AGILE)

Glioblastoma (GBM) represents one of the most promising areas of investigation for Tinostamustine, driven by strong preclinical rationale and a significant unmet medical need. The standard-of-care chemotherapy for newly diagnosed GBM is temozolomide, an alkylating agent whose efficacy is largely dependent on the methylation status of the O6-methylguanine-DNA-methyltransferase (MGMT) gene promoter. Patients with an unmethylated MGMT promoter have high levels of the MGMT repair enzyme, which removes the cytotoxic lesions created by temozolomide, rendering the treatment largely ineffective.[5]

Preclinical studies in GBM models revealed that Tinostamustine's efficacy is independent of MGMT expression status, suggesting it could be a valuable therapeutic option for this resistant patient population.[5] This finding, combined with its excellent ability to penetrate the CNS, has prompted dedicated clinical investigation. A Phase I trial (NCI-2018-00872) is currently underway to specifically evaluate the safety and optimal dose of Tinostamustine, with or without radiation therapy, in patients with newly diagnosed MGMT-unmethylated GBM.[37]

Signifying a major strategic commitment to this indication, Purdue Pharma has entered into an agreement to include Tinostamustine in the GBM AGILE (Glioblastoma Adaptive Global Innovative Learning Environment) trial.[21] GBM AGILE is a pioneering, international Phase 2/3 adaptive platform trial designed to more efficiently and rapidly evaluate multiple investigational therapies against a common control arm.[21] This innovative trial design can significantly accelerate the clinical development timeline. Within this platform, Tinostamustine is slated to be investigated both as a potential first-line adjuvant therapy following standard surgery and chemoradiation, and as a treatment for patients with recurrent disease.[21] Its inclusion in such a high-profile, cutting-edge trial underscores the strong scientific rationale and high level of interest in its potential to become a transformative treatment for this devastating brain cancer.

Consolidated Safety and Tolerability Profile

Summary of Treatment-Emergent Adverse Events (TEAEs)

The safety and tolerability of Tinostamustine have been characterized across its Phase 1 and 2 clinical trials in both hematologic and solid tumor malignancies. The overall safety profile is consistent across studies and is considered predictable and manageable within the context of treating advanced cancers.[11] Nearly all patients treated with Tinostamustine experience at least one treatment-emergent adverse event (TEAE).[11] The most frequently reported TEAEs are primarily hematological and gastrointestinal in nature.[11]

The most common hematological TEAEs include thrombocytopenia (decreased platelet count), anemia, neutropenia, lymphopenia, and leukopenia.[11] Among gastrointestinal events, nausea and vomiting are very common but are generally reported as mild-to-moderate in intensity and are typically well-managed with standard antiemetic prophylaxis.[12] Other frequently observed adverse events include fatigue and infusion-related reactions.[17] A consolidated view of the most common TEAEs reported in the major clinical studies is provided in Table 4.

Table 4: Consolidated Safety Profile: Common TEAEs (≥30%) Across Key Studies

Treatment-Emergent Adverse EventIncidence (%) in Solid Tumors (NCT03345485)Incidence (%) in Hematologic Malignancies (NCT02576496)
Nausea81.8%(Gastrointestinal AEs: 68.8%)
Vomiting31.8%(Gastrointestinal AEs: 68.8%)
Thrombocytopenia54.5%(Hematological AEs: 56.3%)
Anemia45.5%(Hematological AEs: 56.3%)
Lymphopenia40.9%(Hematological AEs: 56.3%)
Leukopenia31.8%(Hematological AEs: 56.3%)
NeutropeniaNot specified ≥30%(Hematological AEs: 56.3%)
Fatigue36.4%Not specified ≥30%
QTc Prolongation59.1%Not specified

Note: Incidence rates for hematologic malignancies are grouped as reported in the source.

Dose-Limiting Toxicities and Events of Special Interest

While many TEAEs are mild to moderate, certain toxicities have been identified as more severe, dose-limiting, or requiring special monitoring.

  • Thrombocytopenia: Severe (Grade 3 or 4) thrombocytopenia is the most consistently reported dose-limiting toxicity (DLT) across the clinical program. In several instances, it has been the primary reason for dose reductions and treatment discontinuations.[11] The management of this side effect is a key aspect of administering the drug safely.
  • QTc Prolongation: Electrocardiogram (ECG) monitoring has revealed a high incidence of QTc interval prolongation. In the Phase 1 solid tumor study, 59.1% of patients experienced some degree of QTc prolongation.[17] While the majority of these events were determined not to be clinically significant, a Grade 3 QTc prolongation was identified as a DLT in a patient receiving the 100 mg/m² dose, highlighting the potential for cardiac toxicity at higher doses.[17] Consequently, clinical trial protocols for Tinostamustine incorporate strict ECG monitoring schedules and predefined stopping rules for significant QTc changes to mitigate this risk.[38]
  • Fatal Adverse Events: The safety database for Tinostamustine is generally favorable with respect to fatal toxicities. Across the main published trial results, no treatment-related deaths were reported in the Hodgkin's lymphoma or solid tumor cohorts.[11] However, one fatal TEAE, an event of decreased appetite, was reported in a patient with mycosis fungoides in the cutaneous T-cell lymphoma cohort of the NCT02576496 study.[12]

Overall Risk-Benefit Assessment

The consolidated safety data for Tinostamustine indicate a toxicity profile that, while significant, is considered both predictable and manageable within the specialized context of oncology. The primary toxicities—myelosuppression (particularly thrombocytopenia) and cardiac repolarization effects (QTc prolongation)—are well-known potential side effects of both alkylating agents and HDAC inhibitors, making them familiar to practicing oncologists.[11] The clinical trial reports consistently describe the safety profile as manageable, suggesting that with appropriate monitoring and supportive care, the risks can be effectively mitigated.[11]

The proactive management of these risks is evident in the drug's development strategy. For instance, the identification of thrombocytopenia as the key DLT has led to the development of specific dose-modification guidelines. This is further underscored by the filing of a patent for a treatment method that involves adjusting the Tinostamustine dose based on a patient's platelet count, representing a formalized strategy to enhance the drug's safety margin.[53] Similarly, the risk of QTc prolongation is addressed through rigorous ECG monitoring protocols.

When weighed against the potential benefits, particularly for patients with advanced, refractory cancers who have exhausted all standard therapeutic options, the risk-benefit profile appears favorable. For these patient populations, where prognosis is often poor and treatment alternatives are scarce, a manageable and predictable safety profile coupled with signals of clinical efficacy makes Tinostamustine a viable and promising investigational agent.

Regulatory and Intellectual Property Landscape

Orphan Drug Designations

Tinostamustine has been granted special regulatory status by major health authorities in recognition of its potential to treat a rare and life-threatening disease. These designations are intended to facilitate and incentivize the development of drugs for conditions with high unmet medical need.

  • U.S. Food and Drug Administration (FDA): In March 2019, the FDA granted Orphan Drug Designation (ODD) to Tinostamustine for the treatment of T-cell prolymphocytic leukemia (T-PLL).[18] T-PLL is an extremely rare and aggressive T-cell leukemia with very limited effective treatment options and a median survival of approximately one year.[18] The ODD status provides potential benefits such as market exclusivity for seven years upon approval, tax credits for clinical research, and waiver of FDA fees.
  • European Medicines Agency (EMA): Following the FDA's decision, the European Commission, on the recommendation of the EMA's Committee for Orphan Medicinal Products (COMP), granted ODD to Tinostamustine for the treatment of T-PLL on July 27, 2020.[19] The designation was based on the seriousness of the condition, the lack of satisfactory authorized treatments in the European Union, and its rarity, affecting approximately 0.1 in 10,000 people in the EU.[20]

The strategic pursuit and successful acquisition of these orphan designations for T-PLL highlight a focused regulatory strategy. By targeting a rare disease with a dire prognosis and no established standard of care, the developers of Tinostamustine have positioned the drug on a potentially accelerated path toward its first marketing approval. Proving a meaningful clinical benefit in this small, well-defined patient population could provide a faster route to market, from which further development in broader indications can be launched.

Patent Portfolio and Market Exclusivity

The commercial potential of Tinostamustine is protected by a growing portfolio of intellectual property, with patents and patent applications filed to cover its composition of matter, methods of use, and specific treatment regimens. The primary assignee and developer of this intellectual property is Purdue Pharma L.P..[53]

The patent portfolio is strategically focused on securing market exclusivity for the use of Tinostamustine in various oncologic indications. Granted patents and published applications cover its therapeutic use in a range of cancers, reflecting the broad scope of its clinical and preclinical investigation. A key patent also covers a method-of-use for managing the drug's primary toxicity, demonstrating an effort to protect not only the drug itself but also the optimal way to administer it safely. A summary of key patents and applications is provided in Table 5.

Table 5: Summary of Key Intellectual Property for Tinostamustine

Patent/Publication NumberTitle/InventionAssigneeStatusSource(s)
US 12370178Tinostamustine for use in the treatment of t-cell prolymphocytic leukaemiaPurdue Pharma L.P.Grant53
US 11918558Tinostamustine for use in the treatment of T-cell prolymphocytic leukaemiaPurdue Pharma L.P.Grant53
US 12257237Tinostamustine for use in treating sarcomaPurdue Pharma L.P.Grant53
US 12377076Compounds for treating lymphoma or a T-cell malignant diseasePurdue Pharma L.P.Grant53
US 20240261265TINOSTAMUSTINE FOR USE IN TREATING OVARIAN CANCERPurdue Pharma L.P.Application53
US 20240139155COMPOUNDS FOR TREATING TNBCPurdue Pharma L.P.Application53

This intellectual property, combined with the market exclusivity periods afforded by its Orphan Drug Designations, provides a strong foundation for the commercial development of Tinostamustine.

Strategic Analysis and Future Outlook

Competitive Landscape and Positioning

Tinostamustine is being developed to address several areas of significant unmet need in oncology, and its strategic positioning varies depending on the specific indication.

  • T-Cell Lymphomas (T-PLL and CTCL): For T-PLL, the treatment landscape is sparse. The standard of care has been the anti-CD52 antibody alemtuzumab, which can induce high response rates but remissions are often not durable, and relapse is common.[60] For many patients, the only curative option is an allogeneic stem cell transplant, which carries significant risk and is not suitable for all patients.[60] In this context, Tinostamustine, with its novel mechanism of action and potential to overcome resistance, is positioned as a much-needed new therapeutic option for both treatment-naïve and relapsed/refractory patients.[45] Similarly, in CTCL, where it demonstrated a 50% ORR and 7.3-month median PFS, it could compete with or complement other approved HDAC inhibitors (like vorinostat and romidepsin) and emerging therapies for patients who have progressed on prior treatments.[45]
  • Relapsed/Refractory Hodgkin's Lymphoma: The treatment paradigm for R/R HL has been revolutionized by the introduction of the antibody-drug conjugate brentuximab vedotin (BV) and PD-1 checkpoint inhibitors (nivolumab, pembrolizumab).[65] However, a subset of patients still relapse after these novel agents and autologous stem cell transplant (ASCT). Tinostamustine is positioned for this heavily pre-treated population.[13] The observed ORR of 37% in patients who have failed multiple prior lines of therapy, including in some cases BV and checkpoint inhibitors, establishes a clinically relevant niche where few effective options exist.[11]
  • Glioblastoma: Tinostamustine's positioning in GBM is highly specific and strategic. It is being targeted primarily for the patient population with MGMT-unmethylated tumors.[37] This subgroup represents nearly half of all GBM patients and derives little to no benefit from the current standard-of-care chemotherapy, temozolomide.[5] The strong preclinical data showing Tinostamustine's efficacy irrespective of MGMT status, combined with its excellent CNS penetration, positions it not as a direct competitor to temozolomide, but as a potential new standard of care specifically for this large, underserved population.[21] Its inclusion in the GBM AGILE trial for both first-line and recurrent settings further solidifies this targeted strategy.[21]

Future Research Directions and Unanswered Questions

The existing data on Tinostamustine's unique mechanism of action points toward several promising avenues for future research and clinical development, primarily centered on rational combination therapies.

  • Combination with Immunotherapies: The preclinical data demonstrating that Tinostamustine can upregulate CD38 and enhance the efficacy of daratumumab provides a powerful proof of concept for "epigenetic priming".[9] This strategy should be expanded. A Phase 1b study combining Tinostamustine with the PD-1 inhibitor nivolumab in advanced melanoma has already been conducted, showing the combination is safe and resulted in a 54% disease stabilization rate in a predominantly ICI-resistant population.[16] Future trials should explore combinations with checkpoint inhibitors in other indications, such as HL and solid tumors, and with other antibody-based therapies whose targets may be under epigenetic control.
  • Combination with DNA Damage Response (DDR) Inhibitors: Tinostamustine's primary function is to induce extensive DNA damage while simultaneously impairing repair processes.[5] This creates a logical synergy with drugs that target other nodes of the DNA damage response pathway. Preclinical evidence suggests potential synergy with PARP inhibitors, which block a key DNA repair mechanism.[35] Clinical trials combining Tinostamustine with a PARP inhibitor in homologous recombination-deficient tumors (e.g., certain ovarian or breast cancers) would be a highly rational next step.
  • Combination with Proteasome Inhibitors: The specific induction of the UPR via HDAC6 inhibition provides a clear mechanistic rationale for combining Tinostamustine with proteasome inhibitors in multiple myeloma.[1] While strong in vitro synergy has been shown, this combination needs to be validated in clinical trials to assess its safety and efficacy in R/R multiple myeloma patients.
  • Biomarker Development: A key unanswered question is which patients are most likely to respond to Tinostamustine. Beyond MGMT status in GBM, there is a need to identify predictive biomarkers of response. Future clinical trials should incorporate robust translational research programs to analyze tumor tissue and circulating tumor DNA (ctDNA) for genetic and epigenetic signatures that correlate with clinical outcomes. Exploratory objectives in ongoing trials are already beginning to address this by profiling tumor DNA, RNA, and methylation patterns.[37]

Concluding Remarks on Therapeutic Potential

Tinostamustine represents a sophisticated example of rational drug design, successfully integrating two distinct, synergistic mechanisms of action into a single molecule. Its development is founded on a compelling scientific hypothesis: that epigenetic modulation can enhance the efficacy of DNA-damaging chemotherapy. The preclinical and clinical data generated to date largely validate this concept.

While its efficacy as a monotherapy in late-stage, pan-refractory patient populations has been modest in terms of objective response rates, it has consistently demonstrated meaningful clinical benefit by achieving durable disease control in patients with no other viable treatment options. This establishes a clear baseline of biological activity and a favorable risk-benefit profile in high-unmet-need settings.

The ultimate therapeutic potential of Tinostamustine, however, likely extends beyond its use as a single agent. Its true value may lie in two key strategic areas. First, as a potentially practice-changing therapy for specific, molecularly defined patient populations that are poorly served by current standards of care, such as MGMT-unmethylated glioblastoma and rare, aggressive leukemias like T-PLL. Second, and perhaps more broadly, as a versatile and powerful combination partner. Its ability to modulate the cellular environment—by relaxing chromatin, inducing the unfolded protein response, and altering the tumor cell immunophenotype—creates unique vulnerabilities that can be exploited by other therapeutic modalities, including DNA repair inhibitors, proteasome inhibitors, and immunotherapies. With a manageable safety profile and a strong, multi-faceted mechanism of action, Tinostamustine is a promising investigational agent that warrants continued and expanded clinical development.

Works cited

  1. Tinostamustine | C19H28Cl2N4O2 | CID 46836227 - PubChem, accessed October 20, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Tinostamustine
  2. Definition of tinostamustine - NCI Drug Dictionary, accessed October 20, 2025, https://www.cancer.gov/publications/dictionaries/cancer-drug/def/tinostamustine
  3. EDO-S101 (Tinostamustine, CAS Number: 1236199-60-2) | Cayman ..., accessed October 20, 2025, https://www.caymanchem.com/product/23328/edo-s101
  4. Tinostamustine(EDO-S101) | HDAC inhibitor | Mechanism - Selleck Chemicals, accessed October 20, 2025, https://www.selleckchem.com/products/tinostamustine.html
  5. The first-in-class alkylating deacetylase inhibitor molecule tinostamustine shows antitumor effects and is synergistic with radiotherapy in preclinical models of glioblastoma, accessed October 20, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5830080/
  6. The first-in-class alkylating deacetylase inhibitor molecule tinostamustine shows antitumor effects and is synergistic with radiotherapy in preclinical models of glioblastoma - PubMed, accessed October 20, 2025, https://pubmed.ncbi.nlm.nih.gov/29486795/
  7. CAS 1236199-60-2 EDO-S101 - BOC Sciences, accessed October 20, 2025, https://www.bocsci.com/edo-s101-cas-1236199-60-2-item-7339.html
  8. CNS pharmacokinetic analysis of EDO-S101 administered by one hour IV... - ResearchGate, accessed October 20, 2025, https://www.researchgate.net/figure/CNS-pharmacokinetic-analysis-of-EDO-S101-administered-by-one-hour-IV-infusion-in-C57BL-6J_fig2_325800186
  9. Tinostamustine (EDO-S101), an Alkylating Deacetylase Inhibitor, Enhances the Efficacy of Daratumumab in Multiple Myeloma by Upregulation of CD38 and NKG2D Ligands - MDPI, accessed October 20, 2025, https://www.mdpi.com/1422-0067/25/9/4718
  10. Tinostamustine (EDO-S101), an Alkylating Deacetylase Inhibitor, Enhances the Efficacy of Daratumumab in Multiple Myeloma by Upregulation of CD38 and NKG2D Ligands - PubMed, accessed October 20, 2025, https://pubmed.ncbi.nlm.nih.gov/38731936/
  11. Safety and Efficacy of Tinostamustine in a Subpopulation of Patients With Relapsed/Refractory Hodgkin Lymphoma From a Phase I Trial - PubMed Central, accessed October 20, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11609130/
  12. Results from the Cohort Expansion Stage of a Phase I Trial of ..., accessed October 20, 2025, https://www.researchgate.net/publication/376151376_Results_from_the_Cohort_Expansion_Stage_of_a_Phase_I_Trial_of_Tinostamustine_Monotherapy_in_Advanced_Haematological_Malignancies_NCT02576496_Findings_from_a_Subset_of_Patients_with_Cutaneous_T-Cell_Ly
  13. (PDF) Safety and Efficacy of Tinostamustine in a Subpopulation of ..., accessed October 20, 2025, https://www.researchgate.net/publication/386326187_Safety_and_Efficacy_of_Tinostamustine_in_a_Subpopulation_of_Patients_With_RelapsedRefractory_Hodgkin_Lymphoma_From_a_Phase_I_Trial
  14. Safety and Efficacy of Tinostamustine in a Subpopulation of Patients With Relapsed/Refractory Hodgkin Lymphoma From a Phase I Trial - PubMed, accessed October 20, 2025, https://pubmed.ncbi.nlm.nih.gov/39617826/
  15. A phase II study of tinostamustine in patients (pts) with advanced solid tumours., accessed October 20, 2025, https://ascopubs.org/doi/10.1200/JCO.2023.41.16_suppl.3021
  16. A phase II study of tinostamustine in patients (pts) with advanced solid tumours., accessed October 20, 2025, https://www.researchgate.net/publication/371297897_A_phase_II_study_of_tinostamustine_in_patients_pts_with_advanced_solid_tumours
  17. Abstract CT023: Dose escalation of tinostamustine in patients with advanced solid tumors, accessed October 20, 2025, https://aacrjournals.org/cancerres/article/79/13_Supplement/CT023/637650/Abstract-CT023-Dose-escalation-of-tinostamustine
  18. Imbrium Therapeutics Announces U.S. FDA Orphan Drug Designation for Tinostamustine for the Treatment of T-cell Prolymphocytic Leukemia, accessed October 20, 2025, https://imbriumthera.com/news/imbrium-therapeutics-announces-u-s-fda-orphan-drug-designation-for-tinostamustine-for-the-treatment-of-t-cell-prolymphocytic-leukemia/
  19. Orphan Drug Designation granted to tinostamustine in EU for very rare form of leukaemia, accessed October 20, 2025, https://pf-media.co.uk/news/orphan-drug-designation-granted-to-tinostamustine-in-eu-for-very-rare-form-of-leukaemia/
  20. Public summary of opinion on orphan designation Tinostamustine ..., accessed October 20, 2025, https://www.ema.europa.eu/en/documents/orphan-designation/eu3202307-public-summary-opinion-orphan-designation-tinostamustine-treatment-t-cell-prolymphocytic-leukaemia_en.pdf
  21. Investigational Drug Tinostamustine to be Included in GBM AGILE Clinical Trial for Glioblastoma - Purdue Pharma, accessed October 20, 2025, https://www.purduepharma.com/news/2025/09/17/investigational-drug-tinostamustine-to-be-included-in-gbm-agile-clinical-trial-for-glioblastoma/
  22. Tinostamustine | MedPath, accessed October 20, 2025, https://trial.medpath.com/drug/5a1b1bc79a5de239/tinostamustine
  23. Mundipharma EDO - Tinostamustine - AdisInsight - Springer, accessed October 20, 2025, https://adisinsight.springer.com/drugs/800045056
  24. STATEMENT ON A NONPROPRIETARY NAME ADOPTED BY THE USAN COUNCIL USAN (EF-22) TINOSTAMUSTINE PRONUNCIATION tye” noe sta mus T, accessed October 20, 2025, https://searchusan.ama-assn.org/usan/documentDownload?uri=/unstructured/binary/usan/tinostamustine.pdf
  25. Tinostamustine free base | CAS#1236199-60-2 - MedKoo Biosciences, accessed October 20, 2025, https://www.medkoo.com/products/33278
  26. Databases : Metabolite Database - Metabolomics Workbench, accessed October 20, 2025, https://www.metabolomicsworkbench.org/data/StructureData.php?RegNo=155309
  27. TINOSTAMUSTINE - precisionFDA, accessed October 20, 2025, https://precision.fda.gov/ginas/app/ui/substances/13124280-6c36-4954-87f2-639ea17e06c9
  28. Tinostamustine (EDO-S101) and Its Combination with Celecoxib or Temozolomide as a Therapeutic Option for Adult-Type Diffuse Gliomas - MDPI, accessed October 20, 2025, https://www.mdpi.com/1422-0067/26/2/661
  29. NCT03345485 | Study of the Safety, Pharmacokinetics and Efficacy of Tinostamustine in Patients With Advanced Solid Tumors. | ClinicalTrials.gov, accessed October 20, 2025, https://www.clinicaltrials.gov/study/NCT03345485
  30. Study of the Safety, Pharmacokinetics and Efficacy of Tinostamustine in Patients With Advanced Solid Tumors. - Clinical Trials | Stanford Medicine, accessed October 20, 2025, https://clinicaltrials.stanford.edu/trials/s/NCT03345485.html
  31. Study of the Safety, Pharmacokinetics and Efficacy of EDO-S101, in Patients With Advanced Solid Tumors - Mayo Clinic, accessed October 20, 2025, https://www.mayo.edu/research/clinical-trials/cls-20385842
  32. Tinostamustine (EDO-S101) | HDAC Inhibitor - MedchemExpress.com, accessed October 20, 2025, https://www.medchemexpress.com/tinostamustine.html
  33. View of Epigenetic approaches. Histone deacetylase inhibitors: vorinostat. A new treatment option for advanced cutaneous T-cell lymphoma - PAGEPress, accessed October 20, 2025, https://www.pagepress.org/journals/hmr/article/view/525/604
  34. The first-in-class alkylating deacetylase inhibitor molecule ..., accessed October 20, 2025, https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5830080/
  35. A Phase 1/2 Study to Investigate the Safety, Pharmacokinetics and Efficacy of Tinostamustine, a First-in-Class Alkylating Histon - Clinical Trials, accessed October 20, 2025, https://cdn.clinicaltrials.gov/large-docs/85/NCT03345485/Prot_001.pdf
  36. The first-in-class alkylating deacetylase inhibitor molecule tinostamustine shows antitumor effects and is synergistic with radiotherapy in preclinical models of glioblastoma - ResearchGate, accessed October 20, 2025, https://www.researchgate.net/publication/323433369_The_first-in-class_alkylating_deacetylase_inhibitor_molecule_tinostamustine_shows_antitumor_effects_and_is_synergistic_with_radiotherapy_in_preclinical_models_of_glioblastoma
  37. Tinostamustine with or without Radiation Therapy in Treating Patients with Newly Diagnosed MGMT-Unmethylated Glioblastoma - NCI, accessed October 20, 2025, https://www.cancer.gov/research/participate/clinical-trials-search/v?id=NCI-2018-00872
  38. A Phase 1/2 Study to Investigate the Safety, Pharmacokinetics and Efficacy of Tinostamustine, a First-in-Class Alkylating Histon - ClinicalTrials.gov, accessed October 20, 2025, https://cdn.clinicaltrials.gov/large-docs/85/NCT03345485/Prot_002.pdf
  39. Tinostamustine (EDO-S101), an Alkylating Deacetylase Inhibitor, Enhances the Efficacy of Daratumumab in Multiple Myeloma by Upregulation of CD38 and NKG2D Ligands - PMC, accessed October 20, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11083018/
  40. Study of Tinostamustine, First-in-Class Alkylating HDACi Fusion Molecule, in Relapsed/Refractory Hematologic Malignancies - NCI, accessed October 20, 2025, https://www.cancer.gov/research/participate/clinical-trials-search/v?id=NCI-2016-00310
  41. NCT02576496 | Study of Tinostamustine, First-in-Class Alkylating HDACi Fusion Molecule, in Relapsed/Refractory Hematologic Malignancies | ClinicalTrials.gov, accessed October 20, 2025, https://www.clinicaltrials.gov/study/NCT02576496
  42. Investigational Drug Tinostamustine to be Included in GBM AGILE Clinical Trial for Glioblastoma - Business Wire, accessed October 20, 2025, https://www.businesswire.com/news/home/20250917676784/en/Investigational-Drug-Tinostamustine-to-be-Included-in-GBM-AGILE-Clinical-Trial-for-Glioblastoma
  43. Study to Investigate the Safety, Pharmacokinetic Profiles and the Efficacy of Tinostamustine, a First-in-Class Alkylating Histone Deacetylase Inhibition Fusion Molecule, in Relapsed/Refractory Hematologic Malignancies - Mayo Clinic, accessed October 20, 2025, https://www.mayo.edu/research/clinical-trials/cls-20193942
  44. Results from the Cohort-Expansion Stage of a Phase I Trial of ..., accessed October 20, 2025, https://www.researchgate.net/publication/376161807_Results_from_the_Cohort-Expansion_Stage_of_a_Phase_I_Trial_of_Tinostamustine_Monotherapy_in_Advanced_Hodgkin_Lymphoma_HL_Non-Hodgkin_Lymphoma_NHL_and_Multiple_Myeloma_MM_NCT02576496
  45. Results from the Cohort Expansion Stage of a Phase I Trial of Tinostamustine Monotherapy in Advanced Haematological Malignancies (NCT02576496): Findings from a Subset of Patients with Cutaneous T-Cell Lymphoma (CTCL) | Blood | American Society of Hematology, accessed October 20, 2025, https://ashpublications.org/blood/article/142/Supplement%201/4432/503332/Results-from-the-Cohort-Expansion-Stage-of-a-Phase
  46. Safety and Efficacy of Tinostamustine in a Subpopulation of Patients With Relapsed/Refractory Hodgkin Lymphoma - PURE.EUR.NL., accessed October 20, 2025, https://pure.eur.nl/files/181383149/Hematological_Oncology_-_2024_-_Sureda_-_Safety_and_Efficacy_of_Tinostamustine_in_a_Subpopulation_of_Patients_With_Relapsed.pdf
  47. Study of Tinostamustine, First-in-Class Alkylating HDACi Fusion Molecule, in Relapsed/Refractory Hematologic Malignancies | Clinical Research Trial Listing - CenterWatch, accessed October 20, 2025, https://www.centerwatch.com/clinical-trials/listings/NCT02576496/study-of-tinostamustine-first-in-class-alkylating-hdaci-fusion-molecule-in-relapsed-refractory-hematologic-malignancies?activelyRecruiting=false&page=150&id=841&slug=bone-neoplasm
  48. Clinical Trial: NCT02576496 - My Cancer Genome, accessed October 20, 2025, https://www.mycancergenome.org/content/clinical_trials/NCT02576496/
  49. Study Details | NCT03345485 | Study of the Safety ... - Clinical Trials, accessed October 20, 2025, https://clinicaltrials.gov/study/NCT03345485
  50. Clinical Trial: NCT03345485 - My Cancer Genome, accessed October 20, 2025, https://www.mycancergenome.org/content/clinical_trials/NCT03345485/
  51. CLINICAL TRIAL PROTOCOL - Clinical Trials, accessed October 20, 2025, https://cdn.clinicaltrials.gov/large-docs/85/NCT03345485/Prot_000.pdf
  52. Safety and Efficacy of Tinostamustine in Heavily Pre-Treated Patients with Relapsed/Refractory (R/R) Hodgkin Lymphoma (HL) | Blood, accessed October 20, 2025, https://ashpublications.org/blood/article/138/Supplement%201/2472/478850/Safety-and-Efficacy-of-Tinostamustine-in-Heavily
  53. Patents Assigned to Purdue Pharma - Justia Patents Search, accessed October 20, 2025, https://patents.justia.com/assignee/purdue-pharma
  54. Novel Agent Granted Orphan Drug Designation for T-Cell ..., accessed October 20, 2025, https://www.targetedonc.com/view/novel-agent-granted-orphan-drug-designation-for-tcell-prolymphocytic-leukemia
  55. Orphan Drug Designation for T-Cell Prolymphocytic Leukemia Agent ..., accessed October 20, 2025, https://checkrare.com/orphan-drug-designation-for-t-cell-prolymphocytic-leukemia-agent/
  56. Tinostamustine granted Orphan Drug Designation by European Commission, accessed October 20, 2025, https://www.europeanpharmaceuticalreview.com/news/125276/tinostamustine-granted-orphan-drug-designation-by-european-commission/
  57. Union Register of medicinal products - Public health - European Commission, accessed October 20, 2025, https://ec.europa.eu/health/documents/community-register/html/o2307.htm
  58. Orphan Drug Designation granted to tinostamustine in EU for very rare form of leukaemia, accessed October 20, 2025, https://www.pharmiweb.com/press-release/2020-08-04/orphan-drug-designation-granted-to-tinostamustine-in-eu-for-very-rare-form-of-leukaemia
  59. Purdue Pharma LP Patent: Treatment of T-cell Prolymphocytic Leukemia with Tinostamustine - Pharmaceutical Technology, accessed October 20, 2025, https://www.pharmaceutical-technology.com/data-insights/purdue-pharma-gets-grant-for-treatment-of-t-cell-prolymphocytic-leukemia-using-tinostamustine/
  60. Advances in Cellular Therapy for T-Cell Prolymphocytic Leukemia - ResearchGate, accessed October 20, 2025, https://www.researchgate.net/publication/358534445_Advances_in_Cellular_Therapy_for_T-Cell_Prolymphocytic_Leukemia
  61. Advances in Cellular Therapy for T-Cell Prolymphocytic Leukemia - Frontiers, accessed October 20, 2025, https://www.frontiersin.org/journals/oncology/articles/10.3389/fonc.2022.781479/full
  62. T-Cell Prolymphocytic Leukemia: An Overview of Current and Future Approaches - PMC, accessed October 20, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7948687/
  63. T-cell prolymphocytic leukemia (T-PLL) | Blood Cancer United, accessed October 20, 2025, https://bloodcancerunited.org/blood-cancer/leukemia/t-cell-prolymphocytic-leukemia-t-pll
  64. T-Cell Prolymphocytic Leukemia: Diagnosis, Pathogenesis, and Treatment - MDPI, accessed October 20, 2025, https://www.mdpi.com/1422-0067/24/15/12106
  65. Paradigm Shifts in Hodgkin Lymphoma Treatment: From Frontline Therapies to Relapsed Disease - ASCO Publications, accessed October 20, 2025, https://ascopubs.org/doi/10.1200/EDBK_433502
  66. Treatment of relapsed or refractory Hodgkin's lymphoma with lenalidomide combined with PD-1 monoclonal antibody: a case report and literature review - Frontiers, accessed October 20, 2025, https://www.frontiersin.org/journals/oncology/articles/10.3389/fonc.2025.1632039/full
  67. Optimizing Treatment for Relapsed/Refractory Classic Hodgkin Lymphoma in the Era of Immunotherapy - MDPI, accessed October 20, 2025, https://www.mdpi.com/2072-6694/15/18/4509
  68. Novel Agents For Relapsed and Refractory Classical Hodgkin Lymphoma: A Review, accessed October 20, 2025, https://www.frontiersin.org/journals/oncology/articles/10.3389/fonc.2022.929012/full
  69. Improved outcomes for relapsed/refractory Hodgkin lymphoma after autologous transplantation in the era of novel agents | Blood | American Society of Hematology, accessed October 20, 2025, https://ashpublications.org/blood/article/141/22/2727/494750/Improved-outcomes-for-relapsed-refractory-Hodgkin
  70. Treatment of relapsed or refractory Hodgkin's lymphoma with lenalidomide combined with PD-1 monoclonal antibody: a case report and literature review - PubMed, accessed October 20, 2025, https://pubmed.ncbi.nlm.nih.gov/40703549/
  71. P12.06.A A PHASE 1 STUDY OF TINOSTAMUSTINE, A NOVEL ALKYLATING DEACETYLASE INHIBITOR, AS ADJUVANT TREATMENT IN NEWLY DIAGNOSED MGMT PROMOTER-UNMETHYLATED GLIOBLASTOMA | Neuro-Oncology | Oxford Academic, accessed October 20, 2025, https://academic.oup.com/neuro-oncology/article/27/Supplement_3/iii116/8272267
  72. Tinostamustine (EDO-S101) and Its Combination with Celecoxib or ..., accessed October 20, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11766231/
  73. Antitumor effects of TINO in glioma cell models: comparison with BDM... - ResearchGate, accessed October 20, 2025, https://www.researchgate.net/figure/Antitumor-effects-of-TINO-in-glioma-cell-models-comparison-with-BDM-and-SAHA-alone-or-in_fig1_323433369
  74. Open-label nonrandomized phase IB study to characterize the safety and recommended dose of tinostamustine in combination with nivolumab in patients with advanced melanoma (ENIgMA) | Request PDF - ResearchGate, accessed October 20, 2025, https://www.researchgate.net/publication/391059638_Open-label_nonrandomized_phase_IB_study_to_characterize_the_safety_and_recommended_dose_of_tinostamustine_in_combination_with_nivolumab_in_patients_with_advanced_melanoma_ENIgMA

Published at: October 20, 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.