MedPath

TW-37 Advanced Drug Monograph

Published:Sep 24, 2025

Generic Name

TW-37

Drug Type

Small Molecule

Chemical Formula

C33H35NO6S

CAS Number

877877-35-5

TW-37: A Comprehensive Preclinical Assessment of a Dual Bcl-2/Mcl-1 Inhibitor

Executive Summary

TW-37 is a potent, nonpeptidic, small-molecule inhibitor of the B-cell lymphoma-2 (Bcl-2) family of anti-apoptotic proteins, developed through a rational, structure-based design strategy. As a dual inhibitor, it demonstrates high affinity for both Myeloid cell leukemia-1 (Mcl-1) and Bcl-2, with a lower but notable affinity for B-cell lymphoma-extra large (Bcl-xL). This profile positions it as a potential tool to address the intrinsic survival mechanisms that drive tumorigenesis and confer resistance to conventional therapies. An extensive body of preclinical research has documented its promising biological activity. Both in vitro and in vivo studies have consistently demonstrated its pro-apoptotic, anti-proliferative, and antiangiogenic effects across a broad spectrum of hematological and solid tumor models, including lymphoma, pancreatic cancer, head and neck cancer, and neuroblastoma. Furthermore, TW-37 has shown significant synergistic potential, enhancing the cytotoxic effects of standard chemotherapeutic regimens such as CHOP (cyclophosphamide, doxorubicin, vincristine, prednisone) and cisplatin.

Despite this robust and compelling preclinical evidence accumulated over more than a decade, TW-37 has not progressed into human clinical trials. This report analyzes the central paradox of TW-37: its profound preclinical promise juxtaposed with its apparent developmental stall. The investigation reveals that this failure to translate from bench to bedside is likely attributable to a confluence of critical factors. Foremost among these is a complete lack of publicly available pharmacokinetic data, including studies on absorption, distribution, metabolism, and excretion (ADME), which represents a fundamental impediment to initiating first-in-human studies. Additionally, the drug's challenging physicochemical properties, particularly its high lipophilicity and poor aqueous solubility, suggest significant formulation and bioavailability hurdles. Finally, TW-37's developmental trajectory coincided with a paradigm shift in the field of Bcl-2 inhibition. The clinical success of Venetoclax, a highly selective Bcl-2 inhibitor with a superior safety profile, established a new benchmark for targeted therapy. In this context, TW-37's broader inhibition profile, including its activity against Bcl-xL—a known mediator of thrombocytopenia—likely rendered it a less attractive clinical candidate due to potential toxicity concerns. Thus, TW-37 stands as a valuable preclinical tool and a case study in drug development, where potent biological activity alone is insufficient to overcome deficits in drug-like properties and a challenging competitive landscape.

Introduction: The Therapeutic Rationale for Targeting Apoptosis in Oncology

Contextual Overview of Apoptosis

Apoptosis, or programmed cell death, is a highly regulated and essential physiological process for maintaining tissue homeostasis, eliminating damaged or infected cells, and guiding normal development.[1] The process is characterized by a distinct set of morphological and biochemical events, including cell shrinkage, chromatin condensation, nuclear fragmentation, and the formation of apoptotic bodies, which are subsequently cleared by phagocytic cells without eliciting an inflammatory response. The intrinsic, or mitochondrial, pathway of apoptosis is a central mechanism for initiating this process in response to cellular stress signals such as DNA damage, growth factor deprivation, or oncogene activation. A critical failure in this pathway is the evasion of apoptosis, which has been established as one of the fundamental hallmarks of cancer. This capability allows malignant cells to survive despite accumulating oncogenic mutations and enduring the cytotoxic insults of anticancer therapies, thereby contributing directly to tumorigenesis, tumor progression, and the development of therapeutic resistance.[2] Consequently, restoring the apoptotic capacity of cancer cells has become a major strategic goal in modern oncology drug development.

The Bcl-2 Family as a Critical Regulator

The intrinsic apoptotic pathway is governed by the B-cell lymphoma-2 (Bcl-2) family of proteins, which act as the master regulators of mitochondrial integrity.[3] These proteins engage in a complex network of interactions to control mitochondrial outer membrane permeabilization (MOMP), the irreversible step that commits a cell to apoptosis. The Bcl-2 family can be functionally categorized into three distinct subgroups based on their structure and role in apoptosis:

  • Anti-apoptotic Proteins: This group includes Bcl-2 itself, Bcl-xL, Mcl-1, Bcl-W, and Bfl-1 (also known as A1). These proteins are the guardians of cell survival. Their primary function is to sequester and inhibit their pro-apoptotic counterparts, thereby preventing the initiation of MOMP. Overexpression of these proteins is a common feature in many cancers and is frequently associated with poor prognosis and resistance to therapy.[4]
  • Pro-apoptotic Effector Proteins: This subgroup consists of Bax (Bcl-2-associated X protein) and Bak (Bcl-2 homologous antagonist/killer). In healthy cells, these proteins exist as inactive monomers. Upon receiving an apoptotic signal, they undergo a conformational change, oligomerize, and insert into the outer mitochondrial membrane, forming pores that lead to MOMP. This results in the release of cytochrome c and other pro-apoptotic factors from the mitochondrial intermembrane space into the cytosol, triggering the activation of the caspase cascade and executing the cell death program.[5]
  • BH3-only Proteins: This is a diverse group of pro-apoptotic proteins that act as the primary sensors of cellular stress. Members include Bid, Bim, Bad, PUMA, and Noxa. They all share a single Bcl-2 homology 3 (BH3) domain, which is essential for their function. Upon activation, they promote apoptosis by either directly activating the effector proteins Bax and Bak (the 'activators' like Bim and Bid) or by binding to and neutralizing the anti-apoptotic proteins (the 'sensitizers' like Bad), thereby liberating the effectors to initiate MOMP.[9]

The balance between these three factions—the pro-survival guardians, the pro-death effectors, and the stress-sensing BH3-only proteins—determines a cell's fate. In cancer, this balance is often tipped decisively toward survival through the overexpression of anti-apoptotic proteins.

BH3 Mimetics as a Therapeutic Strategy

The central role of the Bcl-2 family in controlling apoptosis, and its frequent dysregulation in cancer, provides a compelling rationale for therapeutic intervention. The discovery that the interaction between pro- and anti-apoptotic proteins is mediated by the binding of the BH3 domain of a pro-apoptotic protein into a hydrophobic surface groove on an anti-apoptotic protein led to a breakthrough in rational drug design: the development of "BH3 mimetics".[6] These are small-molecule compounds engineered to structurally mimic the alpha-helical BH3 domain. By fitting into the BH3-binding groove of anti-apoptotic proteins like Bcl-2, Mcl-1, or Bcl-xL, these mimetics competitively disrupt the protein-protein interactions that hold the apoptotic machinery in check.[10] This action effectively unleashes the pro-apoptotic effector proteins, Bax and Bak, to trigger MOMP and induce apoptosis in cancer cells that are "primed for death" by their dependence on overexpressed anti-apoptotic proteins.[8] TW-37 was developed precisely within this therapeutic paradigm as a nonpeptidic small molecule designed to mimic the action of BH3-only proteins and restore the natural process of apoptosis in malignant cells.[6]

Molecular and Physicochemical Profile of TW-37

Chemical Identity and Structure

TW-37 is a well-characterized experimental compound classified as a nonpeptidic small molecule. Its unique identity is established through a comprehensive set of chemical and regulatory identifiers.

  • DrugBank ID: DB17059 [4]
  • CAS Number: 877877-35-5 [14]
  • Type: Small Molecule, nonpeptide [4]
  • IUPAC Name: N-[4-(2-tert-butylphenyl)sulfonylphenyl]-2,3,4-trihydroxy-5-[(2-propan-2-ylphenyl)methyl]benzamide [18]
  • Common Synonyms: The compound is most commonly referred to as TW-37, TW 37, or TW37. It is also known by its systematic chemical names, such as N-(4-((2-tert-butyl)phenyl)sulfonyl)phenyl)-2,3,4-trihydroxy-5-(2-isopropylbenzyl)benzamide.[4]
  • Molecular Formula: C33​H35​NO6​S [10]
  • Structural Representations: For unambiguous identification and use in computational chemistry databases, the following structural identifiers are provided:
  • InChI: InChI=1S/C33H35NO6S/c1-20(2)25-11-7-6-10-21(25)18-22-19-26(30(36)31(37)29(22)35)32(38)34-23-14-16-24(17-15-23)41(39,40)28-13-9-8-12-27(28)33(3,4)5/h6-17,19-20,35-37H,18H2,1-5H3,(H,34,38) [14]
  • InChIKey: PQAPVTKIEGUPRN-UHFFFAOYSA-N [1]
  • SMILES: CC(C)C1=CC=CC=C1CC2=CC(=C(C(=C2O)O)O)C(=O)NC3=CC=C(C=C3)S(=O)(=O)C4=CC=CC=C4C(C)(C)C [18]

Chemically, TW-37 is described as a secondary carboxamide derived from the formal condensation of 2,3,4-trihydroxy-5-[2-(propan-2-yl)benzyl]benzoic acid with 4-[(2-tert-butylphenyl)sulfonyl]aniline. It belongs to the chemical classes of benzamides, sulfones, and benzenetriols.[18]

Physicochemical Properties

The physicochemical properties of a compound are critical determinants of its potential as a therapeutic agent, influencing its formulation, delivery, and pharmacokinetic behavior. The profile of TW-37 reveals several significant challenges for drug development.

  • Molecular Weight: The molecular weight is consistently reported across multiple sources to be approximately 573.7 g/mol.[10]
  • Solubility: TW-37 exhibits very poor aqueous solubility, a characteristic often described as "brick dust" in medicinal chemistry. Reports indicate it is "insoluble in H2O" [21], with a solubility of less than 1 mg/ml [10] and a computed value as low as 0.000596 mg/mL.[4] In contrast, it demonstrates good solubility in organic solvents, particularly dimethyl sulfoxide (DMSO), with reported solubilities of 15 mg/ml, up to 25 mM, and even 100 mM.[10] Its solubility in ethanol is considerably lower but still achievable, especially with gentle warming.[10]
  • Stability and Storage: The compound is stable as a solid for at least four years when stored at -20°C. Stock solutions, typically prepared in DMSO, can be stored at -20°C or below for several months, though aqueous solutions are not recommended for storage beyond one day.[1]
  • Computed Pharmacokinetic Predictors: Computational models provide further insight into the drug-like properties of TW-37. Data from DrugBank indicate a high lipophilicity, with a calculated LogP (a measure of the partition coefficient between octanol and water) ranging from 5.82 to 7.99.[4] The polar surface area is computed to be 123.93 Ų.[4] Based on these properties, TW-37 violates Lipinski's Rule of Five, a widely used guideline for predicting oral bioavailability. Specifically, its molecular weight is greater than 500 Da and its LogP is greater than 5, both of which are associated with poor absorption and permeation.[4]

The collective physicochemical profile of TW-37 presents a formidable challenge for pharmaceutical development. The combination of a high molecular weight, exceptionally high lipophilicity (LogP >> 5), and exceedingly low aqueous solubility are significant liabilities. In drug development, such characteristics often lead to poor absorption from the gastrointestinal tract, resulting in low and highly variable oral bioavailability. Furthermore, these properties complicate the development of stable and safe intravenous formulations, which may require the use of harsh co-solvents or complex delivery systems to achieve therapeutic concentrations. This inherent difficulty in effectively and consistently delivering the molecule to its target site in a biological system represents a major, and potentially insurmountable, hurdle that likely contributed to its stalled clinical development, irrespective of its potent biological activity.

Pharmacodynamics and Cellular Mechanism of Action

Primary Molecular Targets and Binding Profile

TW-37 was rationally developed through a structure-based design strategy to function as a direct inhibitor of the anti-apoptotic members of the Bcl-2 protein family.[5] Its primary molecular targets have been unequivocally identified as Bcl-2, Mcl-1, and Bcl-xL.[4] The binding affinity of TW-37 for these proteins has been quantified in cell-free fluorescence polarization-based binding assays, with results showing remarkable consistency across multiple independent reports. This consistency underscores the well-characterized biochemical profile of the compound.

The core pharmacodynamic feature of TW-37 is its potent and nearly equipotent inhibition of Mcl-1 and Bcl-2, coupled with a weaker, yet still significant, activity against Bcl-xL. This profile distinguishes it from more selective inhibitors that target only a single family member.

Table 4.1: Comparative Binding Affinities (Ki) of TW-37 for Bcl-2 Family Proteins
Target ProteinKi Value (nM)Selectivity Ratio (Bcl-xL/Target)Source(s)
Mcl-1260~4.35
Bcl-2290~3.85
Bcl-xL1110 (1.11 µM)1.05

As shown in Table 4.1, TW-37 binds to Mcl-1 and Bcl-2 with nanomolar affinity (Ki​ = 260 nM and 290 nM, respectively). Its affinity for Bcl-xL is approximately 4-fold lower (Ki​ = 1110 nM).[6] This dual-targeting capability against both Mcl-1 and Bcl-2 is a key characteristic, as overexpression of either protein can confer survival advantages to cancer cells, and Mcl-1 is a known mechanism of resistance to Bcl-2-selective inhibitors.

Molecular Mechanism of Apoptosis Induction

TW-37 exerts its pro-apoptotic effect by functioning as a bona fide BH3 mimetic.[10] Multidimensional NMR spectroscopy studies have confirmed that it binds directly within the hydrophobic BH3-binding groove on the surface of its target proteins, interacting with the same key amino acid residues that are engaged by natural BH3-only proteins like Bim.[6]

The mechanism proceeds through the following steps:

  1. Competitive Binding: TW-37 occupies the BH3-binding groove on anti-apoptotic proteins (Mcl-1, Bcl-2, and Bcl-xL).[9]
  2. Displacement of Pro-apoptotic Proteins: This binding competitively displaces endogenous pro-apoptotic proteins (such as Bim, Bid, and the effector Bax) that were previously sequestered and neutralized by the anti-apoptotic proteins.[6] Co-immunoprecipitation experiments have directly validated this mechanism, showing that TW-37 treatment disrupts the heterodimeric complexes between Bax or truncated-Bid (t-Bid) and the anti-apoptotic proteins. The observed potency for this disruption follows the order of its binding affinity: Mcl-1 > Bcl-2 >> Bcl-xL.[20]
  3. Activation of Apoptotic Cascade: Once liberated, the pro-apoptotic effector proteins (Bax and Bak) are free to oligomerize and trigger MOMP. This leads to mitochondrial depolarization and the release of cytochrome c into the cytosol, which in turn activates the initiator caspase-9 and the executioner caspase-3, culminating in the systematic dismantling of the cell through apoptosis.[5]

Impact on Cell Cycle and Associated Signaling Pathways

Beyond its direct action on the apoptotic machinery, TW-37 elicits profound effects on other critical cellular processes, notably cell cycle progression and survival signaling pathways.

  • S-Phase Cell Cycle Arrest: A unique and consistently reported cellular consequence of TW-37 treatment is the induction of S-phase cell cycle arrest.[9] This effect has been observed in both cancer cells (e.g., pancreatic, head and neck) and endothelial cells. The arrest is underpinned by significant changes in the expression of key cell cycle regulatory genes. Studies have documented the downregulation of positive regulators required for S-phase progression and mitosis, including E2F-1, cdc25A, CDK4, cyclin A, cyclin D1, and cyclin E. Concurrently, TW-37 treatment leads to the upregulation of cell cycle inhibitors, such as p27 and p57.[9] This S-phase arrest mechanism is distinct from the G2/M arrest typically induced by DNA-damaging agents like cisplatin.[12] This mechanistic distinction provides a strong biological rationale for the synergistic anticancer effects observed when TW-37 is combined with cisplatin. By targeting two separate checkpoints in the cell cycle, the combination creates a more comprehensive blockade of the cell's proliferative machinery, making it more difficult for cancer cells to escape and survive. This transforms the empirical observation of synergy into a well-supported, mechanistically rationalized therapeutic strategy.
  • Inhibition of Notch-1 Signaling: In pancreatic cancer models, a novel mechanism of action for TW-37 was identified involving the inhibition of the Notch-1 signaling pathway. Treatment with TW-37 led to the attenuation of Notch-1, its ligand Jagged-1, and downstream target genes such as Hes-1, both in vitro and in vivo.[1] The Notch pathway is a critical driver of proliferation and survival in many cancers, and its inhibition represents an additional, and potentially significant, component of TW-37's overall antitumor activity.

Preclinical Efficacy and Therapeutic Potential

The therapeutic potential of TW-37 has been extensively evaluated in a wide array of preclinical models, demonstrating broad and potent antineoplastic activity as both a single agent and in combination with standard therapies.

In Vitro Antineoplastic Activity Across Tumor Types

In cell culture systems, TW-37 has consistently shown the ability to inhibit proliferation and induce apoptosis in diverse cancer types, often at nanomolar to low-micromolar concentrations.

  • Hematological Malignancies: TW-37 exhibits significant anti-proliferative and pro-apoptotic effects in models of B-cell lymphoma. Notably, this activity was demonstrated in the de novo chemo-resistant WSU-DLCL2 diffuse large cell lymphoma (DLCL) cell line and in primary cells isolated from a lymphoma patient, highlighting its potential to overcome intrinsic resistance mechanisms.[19]
  • Pancreatic Cancer: In a panel of human pancreatic cancer cell lines, including AsPC-1, BxPC-3, and Colo-357, TW-37 induced cell growth inhibition and apoptosis in a dose- and time-dependent manner. This activity was confirmed through multiple assays, including WST-1 cell viability and clonogenic survival assays.[9]
  • Head and Neck Squamous Cell Carcinoma (HNSCC): The compound has shown potent cytotoxicity against HNSCC cell lines (OSCC3, UM-SCC-1, and UM-SCC-74A), with an average half-maximal inhibitory concentration (IC50​) of 0.3 µM. This potency underscores its relevance for a solid tumor type where Bcl-2 family proteins are known to play a pathogenic role.[12]
  • Neuroblastoma: Studies in neuroblastoma cell lines revealed a differential sensitivity based on N-Myc amplification status. N-Myc amplified lines, such as IMR-5 and Kelly, were significantly more sensitive to TW-37 (IC50​ values of 0.28 µM and 0.22 µM, respectively) compared to non-amplified lines. This suggests that N-Myc amplification may serve as a potential biomarker for sensitivity to dual Bcl-2/Mcl-1 inhibition.[15]
  • Colorectal Cancer (CRC): At nanomolar concentrations, TW-37 effectively inhibited cell survival and proliferation and induced caspase-dependent apoptosis in HCT-116 cells and in primary human colon cancer cells. These studies also revealed that TW-37 induces a feedback autophagy response, and that inhibiting this response potentiates TW-37-induced cell death.[11]

The breadth of in vitro activity is summarized in the table below, which consolidates key findings and reinforces the conclusion that TW-37 possesses broad-spectrum anticancer potential.

Table 5.1: Summary of In Vitro Activity of TW-37 in Cancer Cell Lines
Cancer TypeCell Line(s)Key Metric (IC50)Key Cellular EffectsSource(s)
Diffuse Large Cell LymphomaWSU-DLCL2, Primary cellsNot specifiedAnti-proliferative, Pro-apoptotic19
Pancreatic CancerBxPC-3, Colo-357250-750 nM rangeGrowth inhibition, Apoptosis, S-phase arrest9
Head & Neck CancerOSCC3, UM-SCC-1, UM-SCC-74A~0.3 µM (average)Cytotoxicity, S-phase arrest12
Neuroblastoma (N-Myc+)IMR-5, Kelly0.28 µM, 0.22 µMIncreased apoptosis, Reduced proliferation15
Colorectal CancerHCT-116, Primary cells100-300 nM (72h)Inhibition of survival/proliferation, Apoptosis11
Endothelial CellsPrimary HUVEC1.1 µM / 1.8 µMApoptosis, S-phase arrest, Antiangiogenic5

In Vivo Antitumor and Antiangiogenic Efficacy

The promising in vitro results have been successfully translated into animal models, providing a higher level of evidence for the therapeutic potential of TW-37.

  • Antitumor Activity: In a severe combined immunodeficient (SCID) mouse xenograft model using WSU-DLCL2 lymphoma cells, TW-37 demonstrated significant antitumor activity.[20] Similarly, in a Kelly neuroblastoma xenograft model, systemic administration of TW-37 led to a marked decrease in tumor growth and a statistically significant improvement in overall survival.[13]
  • Antiangiogenic Effects: Beyond its direct effects on tumor cells, TW-37 possesses potent antiangiogenic properties. At sub-apoptotic concentrations, it inhibits critical functions of endothelial cells, including cell migration and capillary sprouting.[5] It also blocks the expression of angiogenic chemokines CXCL1 and CXCL8.[5] This in vitro activity was validated in vivo using a murine model of humanized vasculature, where intravenous administration of TW-37 at doses of 3 and 30 mg/kg resulted in a significant decrease in the total number of functional human blood vessels.[5] This dual mechanism—directly killing tumor cells and simultaneously cutting off their blood supply—represents a powerful and comprehensive approach to cancer therapy.

Synergistic Activity in Combination Regimens

A key attribute of a novel anticancer agent is its ability to enhance the efficacy of existing standard-of-care therapies. Preclinical studies have repeatedly shown that TW-37 acts synergistically with conventional chemotherapeutic agents.

  • With CHOP in Lymphoma: In lymphoma models, pre-treatment with TW-37 significantly enhanced the cytotoxic effect of the CHOP regimen in vitro. This synergy was even more pronounced in vivo, where the combination of TW-37 and CHOP led to more complete and sustained tumor inhibition compared to either CHOP or TW-37 administered alone.[6]
  • With Cisplatin in HNSCC: The combination of TW-37 and cisplatin demonstrated enhanced cytotoxic effects against HNSCC cells in vitro. In a corresponding xenograft model, the combination therapy significantly delayed tumor progression, increasing the time to tumor failure (defined as a 4-fold increase in volume) compared to either single-drug treatment.[12]
  • With CDDP/5-FU in Nasopharyngeal Carcinoma (NPC): In NPC models, which are often treated with a combination of cisplatin (CDDP) and 5-Fluorouracil (5-FU), the addition of TW-37 was shown to increase the chemosensitivity of NPC cells and tumors. It prominently promoted apoptosis in NPC cells treated with chemotherapy, suggesting it could be a valuable ancillary drug to overcome chemoresistance.[28]

Pharmacokinetic and Preclinical Safety Profile

Analysis of Available Pharmacokinetic Data

Pharmacokinetics—the study of how the body absorbs, distributes, metabolizes, and excretes a drug (ADME)—is a cornerstone of drug development, providing the essential data needed to design safe and effective dosing regimens for human trials. In the case of TW-37, there is a profound and critical gap in this area.

Authoritative pharmacological databases, including DrugBank, explicitly state that key pharmacokinetic parameters for TW-37 are "Not Available".[4] This includes fundamental data on:

  • Absorption
  • Volume of Distribution
  • Protein Binding
  • Metabolism
  • Route of Elimination
  • Half-life
  • Clearance

A thorough review of the provided preclinical literature confirms this data deficit; none of the studies detail the ADME profile of TW-37.[9] This complete absence of published pharmacokinetic data is the single most significant factor explaining the failure of TW-37 to advance into the clinical phase. In any standard drug development pipeline, demonstrating

in vivo efficacy is followed by a mandatory and comprehensive preclinical ADME and toxicology assessment. This package is non-negotiable, as it is required to establish a safe starting dose, a rational dosing schedule, and to predict potential drug-drug interactions for first-in-human clinical trials. The fact that this information has not been published in the many years since the molecule's discovery strongly suggests one of two possibilities: either these crucial studies were never conducted, or they were conducted and revealed a fatal flaw—such as extremely rapid clearance, the formation of toxic metabolites, or near-zero bioavailability—that was deemed unpublishable and led to the termination of the development program. Therefore, the lack of pharmacokinetic data is not merely a missing piece of information; it is the likely reason for the project's cessation.

Preclinical Toxicology and Safety Assessment

While pharmacokinetic data is absent, the available preclinical studies do provide some insights into the toxicology and safety profile of TW-37, which appears to be favorable within the context of the models tested.

  • Selective Cytotoxicity: A consistent and encouraging finding across multiple studies is the selective cytotoxicity of TW-37 for malignant cells over their normal counterparts. In vitro experiments showed that TW-37 had no significant effect on the viability of normal peripheral blood lymphocytes at concentrations that were highly effective against lymphoma cells.[19] Similarly, treatment with TW-37 was cytotoxic to primary human colon cancer cells but was safe and non-cytotoxic to primary human colon epithelial cells.[11] It also showed no cytotoxic effects on normal fibroblasts at concentrations up to 50 µM, a level far exceeding its effective dose against endothelial cells ( IC50​ of 1.8 µM).[5] This suggests a favorable therapeutic window, where the drug preferentially targets cancer cells, potentially due to their "primed" state of dependence on anti-apoptotic proteins.
  • Systemic Toxicity in Animal Models: The favorable in vitro safety profile was largely recapitulated in in vivo animal studies. In xenograft models of head and neck cancer, treatment with TW-37 effectively inhibited tumor angiogenesis and induced tumor apoptosis "without significant systemic toxicities".[12] This general lack of overt toxicity in animal models is a positive indicator, although it is not a substitute for a formal, regulatory-grade toxicology study.
  • Maximum Tolerated Dose (MTD): The MTD of TW-37 was determined in SCID mice, providing a quantitative measure of its acute toxicity. When administered alone via three intravenous injections, the MTD was 40 mg/kg. When given in combination with the CHOP chemotherapy regimen, the MTD was reduced to 20 mg/kg for three injections, indicating some overlapping toxicity but still demonstrating tolerability in a combination setting.[6]

Developmental Context and Strategic Outlook

Current Development Status

Based on all available evidence, TW-37 is an experimental compound that remains in the preclinical stage of development.[6] Despite the extensive body of research detailing its mechanism of action and anticancer efficacy in laboratory and animal models, there is no indication that it has ever been administered to humans.

It is crucial to clarify a point of potential confusion arising from some databases. Clinical trials listed in association with TW-37 are, upon closer inspection, for the compound Gossypol (also known as AT-101).[36] Gossypol is a natural product from which TW-37 was developed as a "second-generation" benzenesulfonyl derivative through structure-based design.[15] The trials for Gossypol are not trials for TW-37, and there are no registered or completed clinical trials for TW-37 itself.

Comparative Analysis within the Bcl-2 Inhibitor Landscape

The developmental story of TW-37 cannot be understood in isolation. It unfolded during a period of rapid evolution and intense competition in the field of Bcl-2 inhibitor drug discovery, which saw a clear progression from broad-spectrum agents to highly selective molecules.

  • First-Generation Pan-Inhibitors: The first major breakthroughs in the field were ABT-737 and its orally bioavailable successor, Navitoclax (ABT-263). These compounds were potent inhibitors of Bcl-2, Bcl-xL, and Bcl-W. While they demonstrated promising clinical activity in lymphoid malignancies, their development was ultimately hampered by a significant on-target toxicity: dose-limiting thrombocytopenia.[37] This side effect was directly caused by the inhibition of Bcl-xL, which is essential for the survival of platelets. The experience with Navitoclax established Bcl-xL inhibition as a major safety liability for this class of drugs.[8]
  • Second-Generation Selective Inhibitors: The clinical limitations of Navitoclax directly motivated the development of a second generation of inhibitors with improved selectivity. The landmark achievement of this effort was Venetoclax (ABT-199). Through meticulous medicinal chemistry, Venetoclax was engineered to be highly selective for Bcl-2, with over a thousand-fold lower affinity for Bcl-xL.[39] This exquisite selectivity successfully uncoupled the desired anti-leukemic efficacy from the dose-limiting thrombocytopenia. The resulting superior safety profile and profound clinical efficacy led to the approval of Venetoclax for treating chronic lymphocytic leukemia (CLL) and acute myeloid leukemia (AML), fundamentally changing the treatment paradigm for these diseases.[7]
  • Positioning of TW-37: TW-37 occupies an awkward position between these two generations. Its dual inhibition of Bcl-2 and Mcl-1 made it more targeted than Navitoclax in some respects. However, its significant off-target activity against Bcl-xL (Ki​ ~1.1 µM) and its relatively modest selectivity for Bcl-2 over Bcl-xL (~4-fold) placed it at a distinct disadvantage compared to the emerging gold standard of Venetoclax.[6]

Potential Barriers to Clinical Translation

The failure of TW-37 to advance to the clinic can be attributed to several interconnected barriers that likely proved insurmountable.

  • The Pharmacokinetic Hurdle: As previously detailed, the complete lack of published ADME data is the most direct and probable cause for the halt in its development. Without this foundational information, proceeding to human trials would be impossible from both a scientific and a regulatory standpoint.
  • The Toxicity Risk: In the wake of the Navitoclax experience, any new Bcl-2 family inhibitor with significant Bcl-xL activity would face intense regulatory and clinical scrutiny. TW-37's selectivity for Bcl-2 over Bcl-xL is vastly inferior to that of Venetoclax, raising a significant red flag for a high risk of inducing thrombocytopenia. The success of Venetoclax set a new, much higher bar for safety and selectivity that TW-37, by its very design, may not have been able to clear.
  • The Competitive Landscape: By the mid-2010s, when much of the preclinical data on TW-37 was being published, Venetoclax was already demonstrating unprecedented efficacy and a manageable safety profile in clinical trials. The clinical and commercial rationale for advancing another Bcl-2 inhibitor with a potentially worse safety profile and significant formulation challenges would have been exceptionally weak. TW-37 was effectively rendered obsolete by a superior competitor before it could even reach the clinic.
  • The Complexity of Acquired Resistance: While the dual Bcl-2/Mcl-1 inhibition offered by TW-37 is a theoretically sound strategy to preempt or treat resistance, the clinical understanding of resistance mechanisms has also evolved. It is now known that cancer cells can evade Bcl-2 inhibition by upregulating other anti-apoptotic proteins like BCL-XL or by acquiring mutations in downstream effectors like BAX or BAK.[40] The field has largely moved towards addressing resistance through rational combination therapies (e.g., Venetoclax plus a BTK inhibitor in CLL) rather than relying on a single, broader-spectrum agent that carries a higher intrinsic risk of toxicity.
  • Parallel Survival Pathways: The regulation of apoptosis is complex, and other proteins outside the immediate Bcl-2 family can play a role. For instance, some research has proposed that the anti-apoptotic protein Galectin-3 may contribute to the failure of some Bcl-2 inhibitor therapies by providing an alternative survival signal.[2] While this is a speculative point in the specific context of TW-37, it underscores the biological complexity that can confound the efficacy of even potent targeted agents.

Expert Synthesis and Future Directions

Integrated Assessment of TW-37's Therapeutic Promise

TW-37 represents a scientifically significant molecule that embodies both the promise and the pitfalls of modern drug discovery. Its development successfully validated a structure-based design approach, leading to a potent, cell-permeable inhibitor with a clinically relevant dual-targeting profile. However, a holistic assessment reveals a compound whose strengths were ultimately outweighed by fundamental liabilities.

  • Strengths: The primary strength of TW-37 lies in its potent, near-equipotent inhibition of Bcl-2 and Mcl-1. This profile remains therapeutically attractive, as Mcl-1 is a key driver of resistance to Bcl-2-selective agents like Venetoclax. The extensive and compelling preclinical data demonstrating broad efficacy across numerous hematological and solid tumor models, along with proven synergy with standard chemotherapies, firmly established its biological promise as an anticancer agent.
  • Weaknesses and Reasons for Developmental Failure: The downfall of TW-37 as a clinical candidate can be attributed to a convergence of three critical weaknesses. First and foremost is the fatal lack of a viable pharmacokinetic profile, as evidenced by the complete absence of published ADME data. Second, its suboptimal selectivity, particularly its residual activity against Bcl-xL, created an unacceptable risk of thrombocytopenia in a clinical environment where the highly selective Venetoclax had already set a new safety standard. Third, its challenging physicochemical properties—high molecular weight, extreme lipophilicity, and poor aqueous solubility—portended major formulation and bioavailability challenges that would be costly and difficult to overcome.

In conclusion, TW-37 should be regarded as an academically important and potent BH3 mimetic that has served as a valuable preclinical tool for interrogating the biology of apoptosis. However, as a potential therapeutic, it was likely rendered obsolete as a clinical candidate by a combination of intrinsic drug-like property liabilities and the strategic success of a more selective, safer, and ultimately superior competitor.

Recommendations for Future Research

While the resurrection of the TW-37 molecule itself for clinical development is highly unlikely, the knowledge gained from its research provides a valuable roadmap for future drug discovery efforts in this space. The core chemical scaffold of TW-37 could serve as a starting point for a next-generation medicinal chemistry program aimed at designing superior dual Bcl-2/Mcl-1 inhibitors.

The key objectives for such a "TW-37 2.0" program should be to systematically engineer out the molecule's identified flaws:

  1. Enhance Selectivity: The primary goal should be to use detailed structure-activity relationship (SAR) studies to modify the molecule to eliminate or drastically reduce its binding affinity for Bcl-xL. Achieving high selectivity against Bcl-xL is a non-negotiable prerequisite for developing a safe dual inhibitor in the post-Venetoclax era.
  2. Improve Physicochemical Properties: A concurrent effort must focus on improving the drug-like properties of the scaffold. This would involve chemical modifications aimed at reducing lipophilicity (lowering LogP) and increasing aqueous solubility, with the goal of creating a molecule suitable for reliable oral administration and simpler formulation.
  3. Integrate Early ADME/Tox Screening: The most critical lesson from the TW-37 story is the importance of a holistic and integrated development strategy. Any new program must incorporate comprehensive ADME and toxicology screening at the earliest stages of the discovery process. This "fail early, fail fast" approach prevents the investment of significant resources into potent molecules that possess fatal, insurmountable flaws in their drug-like properties.

By following these principles, the scientific legacy of TW-37 can be leveraged to create new therapeutic candidates that retain its potent dual-inhibitory mechanism while possessing the safety and pharmacokinetic profiles necessary for successful clinical translation. The journey of TW-37 thus serves as an important case study for both academic and industrial drug discovery programs, highlighting that the path to a successful medicine requires a delicate balance of potent biology, favorable drug-like properties, and a keen awareness of the evolving clinical and competitive landscape.

Works cited

  1. TW 37 | CAS 877877-35-5 | TW37 - Tocris Bioscience, accessed September 24, 2025, https://www.tocris.com/products/tw-37_4038
  2. Why anti-Bcl-2 clinical trials fail: a solution - PubMed, accessed September 24, 2025, https://pubmed.ncbi.nlm.nih.gov/24338002/
  3. Thirty years of BCL-2: translating cell death discoveries into novel cancer therapies, accessed September 24, 2025, https://www.roswellpark.org/sites/default/files/hernandez_4-25-17_paper_5_nrc.2015.17.pdf
  4. TW-37: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed September 24, 2025, https://go.drugbank.com/drugs/DB17059
  5. TW-37 | Bcl-2 Family Inhibitor - MedchemExpress.com, accessed September 24, 2025, https://www.medchemexpress.com/TW-37.html
  6. Preclinical Studies of TW-37, a New Nonpeptidic Small-Molecule Inhibitor of Bcl-2, in Diffuse Large Cell Lymphoma Xenograft Model Reveal Drug Action on Both Bcl-2 and Mcl-1 | Clinical Cancer Research - AACR Journals, accessed September 24, 2025, https://aacrjournals.org/clincancerres/article/13/7/2226/182369/Preclinical-Studies-of-TW-37-a-New-Nonpeptidic
  7. Mechanisms of action of the BCL-2 inhibitor venetoclax in multiple myeloma: a literature review - Frontiers, accessed September 24, 2025, https://www.frontiersin.org/journals/pharmacology/articles/10.3389/fphar.2023.1291920/full
  8. From basic apoptosis discoveries to advanced selective BCL-2 family inhibitors | Request PDF - ResearchGate, accessed September 24, 2025, https://www.researchgate.net/publication/313800300_From_basic_apoptosis_discoveries_to_advanced_selective_BCL-2_family_inhibitors
  9. TW-37, a Small-Molecule Inhibitor of Bcl-2, Inhibits Cell Growth and Induces Apoptosis in Pancreatic Cancer: Involvement of Notch-1 Signaling Pathway - PMC, accessed September 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3798007/
  10. TW-37 - BPS Bioscience, accessed September 24, 2025, https://bpsbioscience.com/tw-37
  11. The preclinical analysis of TW-37 as a potential anti-colorectal cancer cell agent | PLOS One, accessed September 24, 2025, https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0184501
  12. TW-37, a small molecule inhibitor of Bcl-2, mediates S phase cell cycle arrest and suppresses head and neck tumor angiogenesis - PubMed Central, accessed September 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC2709836/
  13. TW-37, a small-molecule inhibitor of Bcl-2, mediates S-phase cell cycle arrest and suppresses head and neck tumor angiogenesis - ResearchGate, accessed September 24, 2025, https://www.researchgate.net/publication/24283419_TW-37_a_small-molecule_inhibitor_of_Bcl-2_mediates_S-phase_cell_cycle_arrest_and_suppresses_head_and_neck_tumor_angiogenesis
  14. TW-37 (CAS Number: 877877-35-5) | Cayman Chemical, accessed September 24, 2025, https://www.caymanchem.com/product/20999/tw-37
  15. The small molecule Bcl-2/Mcl-1 inhibitor TW-37 shows single-agent cytotoxicity in neuroblastoma cell lines, accessed September 24, 2025, https://d-nb.info/1183252919/34
  16. Full article: Synergistic Effects of TW-37 and ABT-263 on Renal Cell Carcinoma Cells, accessed September 24, 2025, https://www.tandfonline.com/doi/full/10.2147/CMAR.S265788
  17. TW-37 (CAS 877877-35-5) - Abcam, accessed September 24, 2025, https://www.abcam.com/en-us/products/biochemicals/tw-37-ab273490
  18. TW-37 | C33H35NO6S | CID 11455910 - PubChem, accessed September 24, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/tw-37
  19. TW-37 | Bcl-2 inhibitor | Mechanism | Concentration - Selleck Chemicals, accessed September 24, 2025, https://www.selleckchem.com/products/TW-37.html
  20. Preclinical Studies of TW-37, a New Nonpeptidic Small-Molecule Inhibitor of Bcl-2, in Diffuse Large Cell Lymphoma Xenograft Model Reveal Drug Action on Both Bcl-2 and Mcl-1 | Clinical Cancer Research - AACR Journals, accessed September 24, 2025, https://aacrjournals.org/clincancerres/article-abstract/13/7/2226/182369
  21. www.apexbt.com, accessed September 24, 2025, https://www.apexbt.com/tw-37.html
  22. TW 37 - Cancerrxgene - Genomics of Drug Sensitivity in Cancer, accessed September 24, 2025, https://www.cancerrxgene.org/translation/Drug/1149
  23. Preclinical studies of TW-37, a new nonpeptidic small-molecule inhibitor of Bcl-2, in diffuse large cell lymphoma xenograft model reveal drug action on both Bcl-2 and Mcl-1 - PubMed, accessed September 24, 2025, https://pubmed.ncbi.nlm.nih.gov/17404107/
  24. Preclinical Studies of TW-37, a New Nonpeptidic Small-Molecule Inhibitor of Bcl-2, in Diffuse Large Cell Lymphoma Xenograft Mode - AACR Journals, accessed September 24, 2025, https://aacrjournals.org/clincancerres/article-pdf/13/7/2226/1974704/2226.pdf
  25. TW-37 | CAS#877877-35-5 | Bcl-2 inhibitor | MedKoo Biosciences, accessed September 24, 2025, https://www.medkoo.com/products/5934
  26. [PDF] The preclinical analysis of TW-37 as a potential anti-colorectal, accessed September 24, 2025, https://www.semanticscholar.org/paper/The-preclinical-analysis-of-TW-37-as-a-potential-Lei-Ding/a3a803031fd7fb9de57dba26239cb2fe6c93708f
  27. Cytotoxic activity of cisplatin and TW-37, a small-molecule inhibitor... - ResearchGate, accessed September 24, 2025, https://www.researchgate.net/figure/Cytotoxic-activity-of-cisplatin-and-TW-37-a-small-molecule-inhibitor-of-Bcl-2-in_fig1_24283419
  28. Small Molecule Inhibitor TW-37 Is Tolerable and Synergistic With Chemotherapy in Nasopharyngeal Carcinoma - PubMed, accessed September 24, 2025, https://pubmed.ncbi.nlm.nih.gov/28696828/
  29. Population pharmacokinetics: an overview - Australian Prescriber, accessed September 24, 2025, https://australianprescriber.tg.org.au/articles/population-pharmacokinetics-an-overview.html
  30. Pharmacokinetics of drugs: newborn perspective - Bansal - Pediatric Medicine, accessed September 24, 2025, https://pm.amegroups.org/article/view/7227/html
  31. Paediatric pharmacokinetics and drug doses - Australian Prescriber, accessed September 24, 2025, https://australianprescriber.tg.org.au/articles/paediatric-pharmacokinetics-and-drug-doses.html
  32. 37 Pharmacokinetics of intravenous fentanyl in neonates, a systematic review, accessed September 24, 2025, https://experts.mcmaster.ca/display/publication3459009
  33. A phase 0 study of the pharmacokinetics, biodistribution, and dosimetry of 188Re-liposome in patients with metastatic tumors - PMC - PubMed Central, accessed September 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6531516/
  34. Full article: Comparison of putative BH3 mimetics AT-101, HA14-1, sabutoclax and TW-37 with ABT-737 in platelets - Taylor & Francis Online, accessed September 24, 2025, https://www.tandfonline.com/doi/full/10.1080/09537104.2020.1724276
  35. B Cell Lymphoma 2: A Potential Therapeutic Target for Cancer Therapy - MDPI, accessed September 24, 2025, https://www.mdpi.com/1422-0067/22/19/10442
  36. TW-37 - Drug - MedPath, accessed September 24, 2025, https://trial.medpath.com/drug/50781475e8752fea/tw-37
  37. The Development and Current Use of BCL-2 Inhibitors for the ..., accessed September 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5862560/
  38. Current Advances and Future Strategies for BCL-2 Inhibitors: Potent ..., accessed September 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10605442/
  39. BCL2 inhibition: back to the future! | Blood - American Society of Hematology, accessed September 24, 2025, https://ashpublications.org/blood/article/143/18/1787/515839/BCL2-inhibition-back-to-the-future
  40. BCL2 Inhibitors: What's the Latest Research? - Dana-Farber Cancer Institute, accessed September 24, 2025, https://blog.dana-farber.org/insight/2024/03/bcl2-inhibitors-whats-the-latest-research/
  41. Present-Day BCL2 Inhibitor Therapy and Future Therapeutic Directions, accessed September 24, 2025, https://www.bloodcancerstoday.com/post/present-day-bcl2-inhibitor-therapy-and-future-therapeutic-directions

Published at: September 24, 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.