Small Molecule
C39H50N4O6
The compound identified by DrugBank accession number DB01721 is an experimental small molecule that has garnered attention primarily for its structural relationship to Indinavir, an established Human Immunodeficiency Virus (HIV) protease inhibitor.[1] As an analogue of Indinavir, DB01721 has been a subject of research aimed at understanding and potentially overcoming the pervasive challenge of drug resistance in HIV-1 therapy. Its investigation is highlighted by detailed structural and biochemical analyses, particularly its interaction with multi-drug resistant (MDR) variants of the HIV-1 protease enzyme.[2]
The study of DB01721 is emblematic of a critical strategy in antiviral drug development: the modification of existing, clinically successful drugs to address limitations such as emergent viral resistance or suboptimal pharmacological profiles. Indinavir, while a potent therapeutic, has seen its utility diminished by the selection of resistant HIV-1 strains and a demanding clinical regimen.[5] The development of analogues like DB01721, which incorporates specific dual modifications to the Indinavir scaffold, represents a rational medicinal chemistry approach. The evaluation of such analogues against MDR HIV-1 protease aims not only to identify new therapeutic candidates but also to deepen the understanding of structure-activity relationships (SAR) and the molecular mechanisms by which resistance can be circumvented. The detailed characterization of DB01721, particularly the finding that its combined modifications did not lead to improved potency against a resistant protease [3], provides valuable insights into the complexities of inhibitor design and the adaptive capabilities of viral targets. This iterative process of design, synthesis, and testing, even when not yielding a superior drug candidate, contributes significantly to the knowledge base required for developing next-generation antiviral agents.
The primary systematic chemical name for DB01721, consistent with its structural features as an Indinavir analogue with specific modifications, is N-[2-hydroxy-1-indanyl]-5-[(2-tertiarybutylaminocarbonyl)-4(benzodioxol-5-ylmethyl)-piperazino]-4-hydroxy-2-(1-phenylethyl)-pentanamide.[4]
Key database identifiers include:
The consistency between the systematic chemical name and the structural details of the research compound, particularly known as XN2 or XN1336-52 in pivotal studies [4], is crucial for accurate data aggregation. The "benzodioxol-5-ylmethyl" moiety within the systematic name corresponds to the methylenedioxyphenyl group at the P2' position of the inhibitor. Similarly, the "2-(1-phenylethyl)-pentanamide" fragment describes the P1 side chain containing an s-methyl group addition to the benzylic carbon, distinguishing it from the simple phenylmethyl (benzyl) group in Indinavir. This structural correlation across various nomenclature systems and database entries (DrugBank DB01721, PubChem CID 5496642, and the PDB ligand XN2) ensures that this report accurately synthesizes information pertinent to this specific chemical entity.
DB01721 is referenced by a variety of synonyms and research codes across different scientific databases and publications, reflecting its investigational nature. A consolidated list is essential for comprehensive information retrieval:
Table 1: Compound Identifiers for DB01721
Identifier Type | Value | Source/Context |
---|---|---|
Systematic Name | N-[2-hydroxy-1-indanyl]-5-[(2-tertiarybutylaminocarbonyl)-4(benzodioxol-5-ylmethyl)-piperazino]-4-hydroxy-2-(1-phenylethyl)-pentanamide | User Query; Consistent with Schiffer et al. (2002) structure for XN1336-52 |
DrugBank ID | DB01721 | User Query, DrugBank 1 |
PubChem CID | 5496642 | PubChem 7 |
PDB Ligand ID | XN2 | Protein Data Bank (for PDB entry 1k6v) 2 |
PDB Entry (ligand present) | 1k6v | Protein Data Bank 2 |
Research Code (Schiffer et al.) | XN1336-52 | Schiffer et al., Protein Science (2002) 4 |
Other Database ID (e.g., PubChem) | PD060269 | PubChem, Probes-Drugs 7 |
Other Database ID (e.g., PubChem) | NS00071696 | PubChem, COCONUT 7 |
Wikidata ID | Q27467591 | Wikidata 7 |
ChEMBL ID | CHEMBL1257519 | ChEMBL (cross-reference) |
Descriptive Synonym | Analogue of indinavir drug | DrugBank 1 |
This table serves as an essential cross-referencing tool, enabling researchers to accurately identify and consolidate information about DB01721 from diverse scientific literature and databases, thereby preventing confusion that can arise from the use of multiple identifiers for the same experimental compound.
The elemental composition of DB01721 is defined by the molecular formula C39H50N4O6. This formula is consistently reported across major chemical databases, including PubChem [7] and the COCONUT database.[8]
Based on this formula, the computed molecular weight is approximately 670.8 g/mol, as provided by PubChem.[7] More precise calculations yield a monoisotopic mass of 670.37303533 Da (PubChem [7]) and a similar value of 670.37304 Da from the COCONUT database.[8] The Probes-Drugs database also lists a molecular weight of 670.37 for the synonym PD060269.[11] The uniformity of these values across different sources provides high confidence in the basic chemical identity of the compound.
The precise atomic arrangement and bonding of DB01721 are captured by various chemical notation systems:
DB01721 is a chiral molecule possessing multiple stereogenic centers. The precise three-dimensional arrangement of atoms is critical for its biological activity, as interactions with enzyme active sites are highly stereospecific. The InChI string, with its stereochemical layer /t25-,29+,31+,32+,33-,36+/m1/s1, explicitly defines the absolute configuration at each of these chiral centers.[7] Similarly, the full IUPAC name incorporates stereochemical descriptors such as (2S), (1S,2R), etc., to denote these configurations.[7] The canonical SMILES string also encodes this chirality using @ (counter-clockwise) and @@ (clockwise) notations for tetrahedral stereocenters.
The defined stereochemistry of DB01721 is not an arbitrary feature but a fundamental determinant of its interaction with the HIV-1 protease active site. HIV-1 protease, being an enzyme, possesses a chiral active site exquisitely shaped to bind its natural peptide substrates, which also have specific stereochemistry.[5] As a peptidomimetic inhibitor, DB01721 must conform to these stereochemical requirements to achieve effective binding and inhibition. Any deviation in the configuration at one or more of its chiral centers would likely lead to a significant loss of binding affinity and, consequently, biological activity. This underscores the importance of stereocontrolled synthesis and chiral purity in the development and study of such enzyme inhibitors. The specific stereoisomer detailed in the databases and co-crystallized in PDB entry 1k6v is understood to be the pharmacologically relevant form.
A comprehensive set of physicochemical properties for DB01721 has been computed or predicted by various chemoinformatic tools, primarily available through PubChem [7], DrugBank [1], and the COCONUT database.[8] These properties offer insights into its potential behavior as a drug molecule:
The physicochemical profile of DB01721, particularly its high molecular weight and significant lipophilicity leading to violations of common drug-likeness rules like Lipinski's Rule of Five, is a characteristic shared by many HIV protease inhibitors.[5] These properties often arise as a consequence of designing molecules large enough and with appropriate hydrophobic interactions to effectively bind within the spacious and predominantly hydrophobic active site of the HIV-1 protease. While such features can contribute to high binding affinity, they frequently present challenges for drug development, including poor aqueous solubility (as predicted for DB01721 [1]), variable oral bioavailability, and complex pharmacokinetic behavior. Addressing these challenges often requires sophisticated formulation strategies or the co-administration of pharmacokinetic enhancers, although such considerations are beyond the scope of the experimental data available for DB01721. The predicted low water solubility is a direct consequence of its structure and a significant factor that would need to be addressed if further development were pursued.
DB01721 is classified based on its structure, origin, and potential use:
The classification of DB01721 as an "Alpha amino acid amide" and belonging to the subclass of "Amino acids, peptides, and analogues" [8] is particularly informative. This categorization underscores its nature as a peptidomimetic. HIV-1 protease naturally cleaves specific peptide sequences within the viral Gag and Gag-Pol polyproteins.[1] Peptidomimetic inhibitors like DB01721 are designed to mimic these natural substrates, thereby fitting into the enzyme's active site and blocking its function. The presence of amide bonds and structural motifs that resemble amino acid side chains allows these inhibitors to make critical interactions with the various subsites (S1, S2, S1', S2', etc.) of the protease active site. This rational design approach, while effective for achieving potent inhibition, often contributes to the larger molecular size and physicochemical properties (e.g., borderline "drug-likeness") commonly observed in this class of antiviral agents.
The primary pharmacological action of DB01721 is the inhibition of the Human Immunodeficiency Virus type 1 (HIV-1) protease.[1] This viral enzyme plays an indispensable role in the HIV-1 replication cycle. HIV-1 protease is responsible for the post-translational cleavage of the viral Gag and Gag-Pol polyprotein precursors into individual, mature structural proteins and essential viral enzymes (such as reverse transcriptase, integrase, and the protease itself).[1] This proteolytic processing is a critical step for the assembly of new, infectious virions.
DB01721, by virtue of its structural design as a peptidomimetic, is believed to act as a competitive inhibitor. It binds to the active site of the HIV-1 protease, thereby preventing the enzyme from processing its natural polyprotein substrates. This blockade of proteolytic activity results in the production of immature, non-infectious viral particles, ultimately suppressing viral replication and reducing viral load in an infected individual.[1]
While DrugBank indicates that the specific mechanism of action for DB01721 is "Not Available" [1], this likely refers to a lack of dedicated, in-depth published studies detailing the finer nuances of its enzyme kinetics or interaction dynamics beyond its established role as an Indinavir analogue and its direct binding to the protease active site. The structural data from PDB entry 1k6v, where DB01721 (as XN2) is co-crystallized within the HIV-1 protease active site [2], along with its design as an analogue of Indinavir (a well-characterized HIV protease inhibitor [6]), provides compelling evidence for its function as a competitive inhibitor of this viral enzyme. For the purposes of this report, its mechanism is confidently defined by this inhibitory action on HIV-1 protease.
The direct molecular target of DB01721 is the protease enzyme domain which is encoded within the larger Gag-Pol polyprotein of HIV-1.[1] The Gag-Pol polyprotein (a representative UniProt ID being P35963 [1]) serves as a precursor that must be precisely cleaved by the viral protease to liberate the functional components of the virus.
HIV-1 protease is classified as an aspartic protease (or aspartyl protease). Its catalytic activity relies on a pair of conserved aspartic acid residues (typically Asp25 from each monomer of the homodimeric enzyme) located at the enzyme's active site.[1] These residues are crucial for the hydrolysis of peptide bonds in the viral polyproteins. DB01721 is designed to interact with this active site, thereby obstructing its enzymatic function.
Experimentally determined, specific pharmacodynamic parameters for DB01721 are not available in the provided DrugBank summary.[1] However, as an HIV-1 protease inhibitor, its principal pharmacodynamic effect would be the dose-dependent suppression of HIV-1 replication. In a clinical or in vivo setting, this would ideally translate to a measurable reduction in plasma HIV RNA levels (viral load).
The potency of this antiviral effect is typically quantified by parameters such as the half-maximal inhibitory concentration (IC50) or half-maximal effective concentration (EC50) against viral replication in cell-based assays, or by the inhibition constant (Ki) in cell-free enzymatic assays against purified protease. For DB01721 (investigated under the research code XN2 or XN1336-52), IC50 values against purified HIV-1 protease (both wild-type and a multi-drug resistant variant) and dissociation constants (Kd) have been reported in the study by Schiffer C.A. et al. (2002).[4] These specific activity values are detailed in Section IV of this report.
No experimental pharmacokinetic data for DB01721 are available from the provided research snippets. However, DrugBank [1] furnishes a comprehensive suite of in silico predicted ADMET (Absorption, Distribution, Metabolism, Excretion, Toxicity) properties, which offer preliminary insights into its potential disposition and safety profile:
Table 2: Predicted ADMET Properties of DB01721 (from DrugBank [1])
ADMET Parameter | Predicted Value/Classification | Probability (if available) |
---|---|---|
Absorption & Distribution | ||
Water Solubility | 0.0233 mg/mL (Low) | |
LogP (Octanol/Water Partition Coeff.) | 4.37 / 3.94 | |
Human Intestinal Absorption (HIA) | 0.5756 (Moderate to Good) | |
Blood Brain Barrier (BBB) Permeability | 0.9872 (Predicted to Cross) | |
Caco-2 Permeability | 0.7597 (Permeable) | |
Metabolism & Excretion | ||
P-glycoprotein (P-gp) Substrate | Yes | 0.9193 |
P-gp Inhibitor I | Yes | 0.5826 |
P-gp Inhibitor II | No | 0.8226 |
CYP2C9 Inhibitor | Yes (Predicted) | |
CYP2D6 Inhibitor | Yes (Predicted) | |
CYP3A4 Inhibitor | Yes (Predicted) | |
CYP3A4 Substrate | Yes (Predicted) | |
CYP450 Inhibitory Promiscuity | Low | 0.8294 |
Renal Organic Cation Transporter (OCT2) | Non-inhibitor | 0.8833 |
Biodegradation | Not readily biodegradable | 0.9718 |
Toxicity | ||
Ames Test | Non-AMES toxic | 0.715 |
Carcinogenicity | Non-carcinogen | 0.8739 |
Rat Acute Toxicity (LD50, mol/kg) | 2.5504 | |
hERG Inhibition (Predictor I) | Weak inhibitor | 0.9684 |
hERG Inhibition (Predictor II) | Inhibitor | 0.5234 |
These predicted ADMET properties, while computational, provide an initial framework for assessing the potential pharmacokinetic behavior and safety liabilities of DB01721. For instance, the predicted status as a P-glycoprotein substrate and inhibitor, along with the predicted inhibition of key cytochrome P450 enzymes (notably CYP3A4), flags a high potential for drug-drug interactions. This is a common characteristic of HIV protease inhibitors; Indinavir itself is a known CYP3A4 substrate and inhibitor, leading to a complex interaction profile with co-administered medications.[14] If DB01721 were to share this profile, its clinical use would necessitate careful management of concomitant therapies. Furthermore, its status as a P-gp substrate could influence its intracellular accumulation in target cells and its passage across biological barriers, potentially impacting its efficacy and distribution.
The prediction of hERG (human Ether-à-go-go-Related Gene) potassium channel inhibition, even if characterized as "weak" by one predictor model [1], constitutes a significant safety concern. hERG channel blockade is mechanistically linked to QT interval prolongation on the electrocardiogram, which can increase the risk of potentially fatal cardiac arrhythmias such as Torsades de Pointes. This is a common cause of drug candidate failure during preclinical or clinical development, and any hERG liability for DB01721 would require thorough experimental investigation. These predictive data collectively highlight that while an analogue might be designed for improved target potency, its overall pharmacokinetic and safety profile remains a critical determinant of its ultimate therapeutic viability.
DB01721, also identified by research codes XN2 and XN1336-52, was developed and investigated as a structural analogue of Indinavir.[1] Indinavir (Crixivan®) is a potent peptidomimetic inhibitor of HIV-1 protease that was approved for the treatment of HIV/AIDS.[6] However, its clinical utility has been hampered by factors including the development of viral resistance, a demanding dosing schedule requiring significant water intake to prevent nephrolithiasis, and various metabolic side effects.[6] The rationale behind developing Indinavir analogues, such as DB01721, is to address these limitations by creating new chemical entities with potentially improved characteristics. These improvements could include enhanced potency against drug-resistant HIV-1 strains, more favorable pharmacokinetic properties (e.g., increased metabolic stability, better oral bioavailability), or a reduced toxicity profile.[5] The research involving DB01721 specifically focused on its interaction with a multi-drug resistant form of HIV-1 protease.
The most definitive research characterizing DB01721 is documented in the study by King N.M., Schiffer C.A., and colleagues, published in Protein Science in 2002, and associated with the Protein Data Bank (PDB) entry 1k6v.[3] This work provides a detailed structural and biochemical analysis of DB01721 (referred to as XN2 in the PDB and XN1336-52 in the paper) in complex with a multi-drug-resistant (MDR) variant of HIV-1 protease.
Compound | Target Protease | IC50 (µM) | Kd (M) |
---|---|---|---|
Indinavir | Wild-Type | Not explicitly stated in table format for IC50, but generally highly potent | 8.6×10−10 |
Indinavir | 3X-Resistant (L63P/V82T/I84V) | 0.47 | 3.3×10−8 |
XN1336-51 (P1 S-methylbenzyl) | Wild-Type | Not explicitly stated in table format for IC50 | 1.2×10−9 |
XN1336-51 (P1 S-methylbenzyl) | 3X-Resistant (L63P/V82T/I84V) | 0.40 | 2.4×10−8 |
807-29-4 (P2' methylenedioxyphenyl) | Wild-Type | Not explicitly stated in table format for IC50 | 5.3×10−10 |
807-29-4 (P2' methylenedioxyphenyl) | 3X-Resistant (L63P/V82T/I84V) | 0.31 | 4.4×10−8 |
DB01721 (XN2/XN1336-52; P1+P2' mods) | Wild-Type | Not explicitly stated in table format for IC50 | 1.5×10−9 |
DB01721 (XN2/XN1336-52; P1+P2' mods) | 3X-Resistant (L63P/V82T/I84V) | 1.2 | 1.2×10−7 |
The data clearly demonstrate that while the singly modified analogues (XN1336-51 and 807-29-4) maintained or slightly improved potency against the 3X-resistant protease compared to Indinavir, the dually modified DB01721 (XN1336-52) exhibited significantly *reduced* potency. Its <span class="math-inline">IC\_\{50\}</span> against the 3X-resistant protease was 1.2 µM, which is 2.5 to 3.8 times weaker than Indinavir and the singly modified compounds. Similarly, its binding affinity (<span class="math-inline">K\_d</span>) for the 3X-resistant protease was also notably poorer. Against the wild-type protease, DB01721 (<span class="math-inline">K\_d \= 1\.5 \\times 10^\{\-9\}</span> M) was less potent than Indinavir (<span class="math-inline">K\_d \= 8\.6 \\times 10^\{\-10\}</span> M) and the P2'-modified analogue 807-29-4 (<span class="math-inline">K\_d \= 5\.3 \\times 10^\{\-10\}</span> M).
The synthesis of DB01721 (XN1336-52) was reported by Schiffer et al. (2002).[4] The key steps involved:
The syntheses of the singly modified analogues XN1336-51 (P1 S-methylbenzyl only) and 807-29-4 (P2' methylenedioxyphenyl only) followed analogous synthetic strategies, utilizing the appropriate aldehyde (pyridine-3-carboxaldehyde for XN1336-51, and piperonal for 807-29-4 with the original Indinavir P1 precursor) in the reductive alkylation step.[4]
The development of DB01721 occurred within a broader context of research aimed at optimizing Indinavir and other HIV protease inhibitors. Several distinct strategies have been pursued:
These varied research efforts underscore the multifaceted approaches taken to refine and build upon the initial success of HIV protease inhibitors like Indinavir, addressing challenges such as resistance, pharmacokinetics, and exploring new therapeutic applications.
DB01721 is classified as an experimental compound.[1] The primary and most detailed characterization of this molecule in the provided research materials dates back to the 2002 publication by Schiffer C.A. et al..[4] There is no indication from the available information that DB01721 has progressed into formal clinical development or received any regulatory approvals.
There are no clinical trials listed for DB01721 in Phases 0, 1, 2, 3, or 4 in the DrugBank database.[1] Searches using other identifiers such as PD060269 or PubChem CID 5496642 in clinical trial registries did not yield any specific trials for this compound.[32]
The absence of clinical trial data, combined with the 2002 publication date of its primary characterization which demonstrated that the dual modifications in DB01721 led to reduced potency against the MDR HIV-1 protease [4], strongly suggests that this specific compound did not advance into clinical development. It is more likely that DB01721 remained a research tool or a case study that contributed to the understanding of inhibitor design principles rather than progressing as a viable drug candidate itself. Compounds exhibiting a less favorable preclinical profile, particularly in terms of target potency compared to existing agents or simpler analogues, are typically not selected for the resource-intensive process of clinical evaluation.
The predicted physical state of DB01721 at standard conditions is solid, according to DrugBank.[1]
DrugBank lists zero patents specifically for DB01721.1 However, the field of Indinavir and its analogues is extensively covered by patents, primarily assigned to Merck & Co., the original developer of Indinavir. Indinavir itself was patented in 1991.6
The synthesis of the P2' methylenedioxyphenyl intermediate, which is a structural component of DB01721 (via analogue 807-29-4), was based on a Merck patent (US 5,413,999A, as cited in the context of Indinavir preparation 17) according to the Schiffer et al. (2002) paper.4 This patent covers Indinavir and related HIV protease inhibitors. Therefore, while DB01721 as a distinct entity may not be individually patented or may have been covered under broader Markush claims of existing patents, its structural motifs and the general class of Indinavir analogues fall within a well-established patent landscape for HIV protease inhibitors.26
Specific information regarding commercial chemical suppliers for DB01721 (PubChem CID 5496642) is not explicitly detailed in the provided research snippets. General chemical supplier advertisements, such as BOC Sciences and TCI America, were noted in one snippet [37], but these were in the context of a different chemical compound (5-hydroxypentan-2-one) and do not confirm availability of DB01721. Given its status as an experimental compound primarily characterized in a 2002 research paper, DB01721 is unlikely to be a readily available catalog chemical. Its acquisition would most likely require custom synthesis. Generic supplier information for peptides or other chemicals [38] is not directly applicable.
The PubChem entry for CID 5496642 (DB01721) includes sections for "Biological Test Results" and "Interactions and Pathways".[7] A general search for bioactivity related to synonyms of DB01721 (PD060269, XN2, 1k6v) and HIV yielded some contextual results.[10] One result referred to the general challenges in HIV protease inhibitor design and the promiscuity of early aspartyl protease inhibitors.[40] Another result pertained to Xanthohumol (abbreviated XN), which is a distinct compound and not the XN2 ligand relevant here, highlighting a potential point of confusion if identifiers are not carefully managed.[41]
The most relevant bioactivity information directly linked to DB01721 (CID 5496642) in PubChem is its association as the ligand XN2 in the PDB entry 1k6v.[7] This PDB entry, and the associated publication by Schiffer et al. (2002) [3], remains the primary source of detailed public bioactivity data (i.e., IC50 and Kd values against HIV-1 protease) for this compound. Beyond this specific structural and biochemical study, there is no indication in the provided materials of extensive public bioassay screening data for DB01721. Its characterization appears largely confined to this focused research project aimed at understanding structure-activity relationships in the context of drug resistance.
DB01721, systematically named N-[2-hydroxy-1-indanyl]-5-[(2-tertiarybutylaminocarbonyl)-4(benzodioxol-5-ylmethyl)-piperazino]-4-hydroxy-2-(1-phenylethyl)-pentanamide, is an experimental analogue of the HIV protease inhibitor Indinavir. Its design incorporated two specific structural modifications to the Indinavir scaffold: an S-methyl group at the P1 benzylic position and a methylenedioxyphenyl moiety replacing the P2' pyridine ring.
The principal investigation of DB01721 focused on its efficacy as an inhibitor of HIV-1 protease, with particular emphasis on its activity against a multi-drug resistant variant (L63P/V82T/I84V). The seminal study by Schiffer C.A. et al. (2002), associated with PDB entry 1k6v, revealed that, contrary to the potential expectation of synergistic or additive benefits from combining two individually tolerated modifications, DB01721 exhibited reduced binding affinity and inhibitory potency against both wild-type and the 3X-resistant HIV-1 protease when compared to Indinavir and the corresponding singly modified analogues.[4] Structural analysis from the co-crystal structure (PDB: 1k6v) indicated that the protease active site could not optimally accommodate both modifications simultaneously, leading to this observed lack of synergy.
The study of DB01721 provides a significant case study in the field of structure-based drug design, particularly for HIV-1 protease inhibitors. It underscores the principle that combining structural modifications, even those that individually appear neutral or beneficial, does not invariably lead to an improved compound. The outcome for DB01721 highlights the intricate and often unpredictable nature of protein-ligand interactions, the critical role of protein conformational flexibility and adaptability ("induced fit"), and the formidable challenges posed by viral drug resistance.
This research emphasizes that a nuanced understanding of how multiple modifications interact—both sterically and allosterically—to affect the overall binding free energy is paramount. The "negative data" generated from the DB01721 investigation, i.e., the documented failure to achieve enhanced potency through these combined modifications, is scientifically valuable. It informs the field about specific structural combinations that are counterproductive for this particular scaffold and target, thereby guiding future design efforts away from similar pitfalls and towards more promising strategies. Such findings refine the SAR models for this class of inhibitors and contribute to a more sophisticated approach to overcoming drug resistance.
Given the reported reduced potency in the key published study [4], DB01721 itself is not a strong candidate for further clinical development in its current form. Significant redesign would be necessary to overcome the observed detrimental effects of the combined modifications.
However, the insights gleaned from its detailed structural and biochemical characterization remain valuable for the broader endeavor of HIV protease inhibitor design. Understanding precisely why this specific combination of P1 and P2' modifications failed to enhance (and in fact, diminished) activity can inform the development of novel scaffolds or different combinations of substituents that might better exploit the adaptive capacity of the protease active site. The overarching challenge in HIV therapeutics persists: to design protease inhibitors that are not only highly potent against wild-type virus but also maintain robust activity against a wide spectrum of clinically relevant resistant variants, while simultaneously possessing favorable pharmacokinetic profiles (good oral bioavailability, manageable drug interactions, appropriate half-life) and minimal toxicity. The story of DB01721 contributes a crucial, albeit cautionary, chapter to this ongoing scientific pursuit.
Published at: June 4, 2025
This report is continuously updated as new research emerges.