[HTMC0435 is an orally bioavailable, small molecule, new molecular entity under investigation as a highly selective dual inhibitor of Poly(ADP-ribose) polymerase 1 and 2 (PARP1/2). Developed through a collaboration between Chinese biopharmaceutical firms Shanghai Huilun Life Science & Technology Co. Ltd. and Shanghai Yidian Pharmaceutical Technology Development Co., Ltd., HTMC0435 is strategically positioned within the validated class of PARP inhibitors, which have become a cornerstone of therapy for cancers with deficiencies in DNA damage repair pathways. The current clinical development program is primarily focused on oncology indications with high unmet medical need, with the most advanced data emerging from its evaluation in extensive-stage small cell lung cancer (ES-SCLC).]
[In the Phase 1b/2 clinical trial NCT05728619, HTMC0435 in combination with the alkylating agent temozolomide has demonstrated compelling antitumor activity in heavily pre-treated patients with relapsed/refractory ES-SCLC. The combination yielded an objective response rate (ORR) of 24.5% and a median overall survival (OS) of 12.0 months. A particularly significant finding is the regimen's profound efficacy in the platinum-resistant subgroup—a population with exceptionally poor prognosis—where it achieved a median OS of 12.6 months. This suggests a mechanism of action that may be independent of platinum sensitivity, potentially offering a new standard of care for this difficult-to-treat population. The safety profile of the combination is characterized by predictable and manageable hematologic toxicities, with a notable zero percent discontinuation rate due to treatment-related adverse events, underscoring its favorable therapeutic index.]
[Beyond SCLC, the developers are pursuing a broader strategy, with active Phase 2 trials investigating HTMC0435 in advanced biliary tract cancer and advanced pancreatic cancer, indications where PARP inhibition is biologically rational but less clinically established. The development strategy appears to be centered on securing initial approval in China, while simultaneously building a global presence through data presentations at major international conferences and securing regulatory designations such as Orphan Drug status in the United States. The ultimate trajectory of HTMC0435 will depend on the successful completion of pivotal, randomized Phase 3 trials to confirm these promising early results. Nevertheless, the existing data package positions HTMC0435 as a significant and promising therapeutic agent with the potential to meaningfully impact the treatment landscape for several aggressive malignancies.]
The family of Poly(ADP-ribose) polymerase (PARP) enzymes, particularly the abundant PARP1 and PARP2 isoforms, are critical mediators of cellular homeostasis.[1] These enzymes play a central role in the DNA damage response (DDR), primarily by detecting and signaling the presence of DNA single-strand breaks (SSBs) and recruiting other repair factors to the site of damage. This function is essential for maintaining genomic stability and ensuring cell survival.[1]
The therapeutic exploitation of PARP inhibition in oncology is rooted in the concept of "synthetic lethality." This principle describes a situation where a deficiency in either of two distinct genes or pathways is compatible with cell viability, but the simultaneous loss of both results in cell death.[1][ Many cancers harbor inherent defects in key DNA repair pathways, most notably the Homologous Recombination (HR) pathway, which is responsible for the high-fidelity repair of DNA double-strand breaks (DSBs). Germline or somatic mutations in genes such as]
BRCA1 and BRCA2 are the canonical examples of Homologous Recombination Deficiency (HRD).[1]
In HR-deficient cancer cells, the repair of DSBs is compromised, making the cells highly dependent on other repair mechanisms, including the PARP-mediated SSB repair pathway. The administration of a PARP inhibitor (PARPi) pharmacologically disables this alternative pathway. When SSBs occur, which are frequent events in rapidly dividing cancer cells, PARP inhibition prevents their efficient repair. During DNA replication, these unresolved SSBs are converted into lethal DSBs. In a normal cell with functional HR, these DSBs can be repaired. However, in an HR-deficient cancer cell, the accumulation of irreparable DSBs triggers catastrophic genomic instability and subsequent apoptosis, or programmed cell death.[1]
This elegant, tumor-selective mechanism of action has led to the successful development and approval of several PARP inhibitors, including olaparib, niraparib, rucaparib, and talazoparib. These agents have transformed the standard of care for patients with HRD-positive cancers, with established indications in ovarian, breast, prostate, and pancreatic cancers.[2][ Despite this success, the field continues to face challenges, including the development of acquired resistance to PARP inhibition and the need to expand their utility beyond the canonical]
BRCA-mutated populations. This has spurred the development of next-generation inhibitors and novel combination strategies designed to overcome resistance or induce a state of "BRCAness" in tumors that are not intrinsically HRD.[2]
Within this dynamic therapeutic landscape, HTMC0435 emerges as a novel, investigational new molecular entity (NME).[5] It is characterized as a highly selective, orally bioavailable small molecule designed to inhibit PARP.[6] Its development represents an effort to build upon the established success of the PARPi class while potentially addressing existing limitations or expanding into new therapeutic areas. As an orally administered agent, HTMC0435 offers the potential for convenient, long-term dosing, a key attribute for maintenance therapies or chronic treatment settings in oncology.[5][ This report provides a comprehensive analysis of the available preclinical, chemical, and clinical data for HTMC0435 to evaluate its pharmacological profile, clinical potential, and strategic position within the field of oncology.]
[The fundamental identity of HTMC0435 is defined by its specific chemical and physical characteristics, which are critical for its synthesis, formulation, and preclinical evaluation. As an investigational compound, this information is primarily available from chemical suppliers and regulatory filings.]
The molecule is identified by the Chemical Abstracts Service (CAS) Number 2024616-54-2.[6][ Its full chemical name is]
4-(3-(4-(1H-imidazo[4,5-b]pyridin-5-yl)piperazine-1-carbonyl)-4-fluorobenzyl)phthalazin-1(2H)-one.[6][ This systematic name precisely describes its complex heterocyclic structure. The molecular properties derived from this structure are a]
molecular formula of C26H22FN7O2 and a corresponding molecular weight of 483.51 g/mol.[6]
Physically, HTMC0435 is supplied as a solid powder. For laboratory purposes, its solubility is characterized as 10 mM in dimethyl sulfoxide (DMSO), a common solvent for in vitro assays. Stability data indicates that the solid powder is stable for 12 months when stored at -20°C, while solutions in DMSO should be stored at -80°C for long-term stability.[6][ These properties are summarized in Table 1.]
Table 1: Physicochemical and Pharmacological Properties of HTMC0435
Property | Value | Source(s) |
---|---|---|
Drug Name | HTMC0435 (also HTMC-0435) | 5 |
Drug Class | Antineoplastics, Small molecules | 5 |
Mechanism of Action | Poly(ADP-ribose) polymerase (PARP1/2) inhibitor | 5 |
New Molecular Entity | Yes | 5 |
Chemical Name | 4-(3-(4-(1H-imidazo[4,5-b]pyridin-5-yl)piperazine-1-carbonyl)-4-fluorobenzyl)phthalazin-1(2H)-one | 6 |
CAS Number | 2024616-54-2 | 6 |
Molecular Formula | C26H22FN7O2 | 6 |
Molecular Weight | 483.51 | 6 |
Appearance | Solid | 6 |
Solubility | 10 mM in DMSO | 6 |
HTMC0435 is explicitly defined as a dual inhibitor of both PARP1 and PARP2, the two most well-characterized members of the PARP family involved in DNA repair.[7][ Like other agents in its class, its primary mechanism involves competitive inhibition at the catalytic domain of the PARP enzyme. PARP enzymes utilize nicotinamide adenine dinucleotide (]
NAD+) as a substrate to synthesize and attach polymers of poly(ADP-ribose) (PAR) onto target proteins at sites of DNA damage. This PARylation process serves as a scaffold to recruit downstream DNA repair factors.[1]
By competing with NAD+ for binding to the catalytic site, HTMC0435 blocks this enzymatic activity. This inhibition prevents the formation of PAR polymers, thereby disrupting the recruitment of the repair machinery and leading to the persistence of SSBs.[1] While direct inhibition of catalytic activity is a key component, a more potent mechanism of cytotoxicity for some PARP inhibitors is "PARP trapping." This phenomenon occurs when the inhibitor binds to the enzyme in such a way that it stabilizes the PARP-DNA complex, effectively "trapping" the enzyme on the DNA lesion. These trapped complexes are highly cytotoxic, as they obstruct DNA replication and transcription, leading to the formation of DSBs.[4][ The relative potency of HTMC0435 as a PARP trapper compared to other approved agents is not specified in the available documentation but remains a critical aspect of its pharmacological profile.]
The clinical development strategy for HTMC0435 has prominently featured its combination with the oral alkylating agent temozolomide (TMZ), particularly in the pivotal NCT05728619 trial for SCLC.[10][ This combination is not arbitrary but is founded on a strong and well-understood synergistic mechanism.]
The therapeutic action of TMZ involves the methylation of DNA, primarily at the N7 and O6 positions of guanine and the N3 position of adenine. These DNA adducts distort the DNA helix and, if not repaired, can stall DNA replication and trigger cell death. The PARP-mediated base excision repair (BER) pathway is a primary mechanism by which cells repair the types of DNA damage induced by TMZ. Preclinical studies have shown that combining a PARP inhibitor with TMZ significantly enhances tumor cell killing.[11]
[The sequence of events underlying this synergy is as follows:]
The promising clinical results observed in the NCT05728619 trial, where this combination has demonstrated significant antitumor activity, serve as a powerful clinical validation of this synergistic mechanism.[10]
The development of HTMC0435 is led by a collaboration of Chinese pharmaceutical companies. The originator organization, responsible for the initial discovery and preclinical development of the molecule, is Shanghai Huilun Life Science & Technology Co. Ltd., also referred to as Shanghai Huilun Pharmaceutical.[5]
The clinical-stage development and execution of the trial program are being conducted jointly by Shanghai Huilun and Shanghai Yidian Pharmaceutical Technology Development Co., Ltd..[5] Both are listed as active organizations in the drug's development pipeline.[7] Notably, Shanghai Yidian Pharmaceutical is the lead sponsor for several key clinical trials, including the pivotal Phase 1b/2 study in SCLC (NCT05728619).[12]
This corporate structure points to a focused, China-centric initial development strategy. The primary clinical trials are registered and conducted within China, leveraging the domestic clinical research infrastructure and patient populations to efficiently generate data.[7][ This approach is common for emerging Chinese biopharmaceutical firms, allowing them to advance assets rapidly toward potential approval by the National Medical Products Administration (NMPA) of China.]
However, the development strategy also exhibits clear signs of global ambition. The presentation of key clinical data at the American Society of Clinical Oncology (ASCO) Annual Meeting is a deliberate effort to engage with the international oncology community, seek external validation of their findings, and attract potential partners for ex-China development and commercialization.[5] Furthermore, the pursuit and attainment of regulatory designations from the U.S. Food and Drug Administration (FDA), such as Orphan Drug status, and the initiation of preclinical work in the United States are concrete steps toward a global regulatory pathway.[7][ This dual-track approach—securing a domestic foundation while simultaneously building a global profile—suggests a sophisticated and forward-looking strategy to maximize the asset's worldwide potential.]
The clinical development plan for HTMC0435 is ambitious, targeting multiple solid tumor types where PARP inhibition is biologically rational. The program is advancing on several fronts, with the highest development phase currently being Phase 2.[5]
The lead indication, with the most mature and publicly available data, is Small Cell Lung Cancer, which is being investigated in a Phase 1/2 trial.[5][ Beyond this, the program has expanded into several other malignancies, with multiple Phase 2 trials actively recruiting patients in China as of late 2023. These include:]
Earlier-stage development activities further underscore the breadth of the program. Investigational New Drug (IND) applications have been approved in China for Castration-Resistant Prostate Cancer and Advanced Pancreatic Adenocarcinoma, allowing clinical trials to commence in these indications. IND applications have also been filed for Ovarian Cancer and Prostatic Cancer, signaling intent to move into these classic PARP inhibitor-sensitive tumor types.[7][ An overview of the clinical pipeline is provided in Table 2.]
Table 2: Summary of Ongoing and Planned Clinical Trials for HTMC0435
Trial Identifier | Indication | Phase | Status (as of late 2023/early 2024) | Country/Location | Key Intervention |
---|---|---|---|---|---|
NCT05728619 | Extensive-Stage Small Cell Lung Cancer (ES-SCLC) | 1/2 | Active, Recruiting | China | HTMC0435 + Temozolomide |
CTR20230615 | Advanced Biliary Tract Cancer | 2 | Recruiting | China | HTMC0435 Monotherapy |
CTR20230616 | Advanced Pancreatic Cancer | 2 | Recruiting | China | HTMC0435 Monotherapy |
CTR20233204 | Advanced Solid Tumors | 2 | Recruiting | China | HTMC0435 + Temozolomide |
CTR20200573 | Advanced Malignant Solid Tumors | 1/2 | Initiated (May 2020) | China | HTMC0435 Monotherapy |
N/A | Castration-Resistant Prostatic Cancer | IND Approval | N/A | China | N/A |
N/A | Advanced Pancreatic Adenocarcinoma | IND Approval | N/A | China | N/A |
N/A | Ovarian Cancer | IND Application | N/A | China | N/A |
N/A | Prostatic Cancer | IND Application | N/A | China | N/A |
N/A | Small Cell Lung Cancer | Preclinical | N/A | United States | N/A |
HTMC0435 is confirmed to be a New Molecular Entity (NME), a critical designation that ensures it is not a derivative of an existing approved drug and is eligible for a full period of market and data exclusivity upon approval.[5]
The drug has also received Orphan Drug Designation from the FDA in the United States.[7] This status is granted to drugs intended to treat rare diseases (affecting fewer than 200,000 people in the U.S.) and provides significant development incentives, including a seven-year period of market exclusivity for the approved indication, tax credits for clinical research, and a waiver of the Prescription Drug User Fee Act (PDUFA) fee. This designation not only provides commercial advantages but also validates the drug's potential to address a significant unmet medical need. It should be noted that one source indicates no orphan status, but the more specific information from other sources points to a U.S. designation.[5]
Extensive-stage small cell lung cancer (ES-SCLC) is one of the most aggressive and lethal solid tumors, characterized by rapid growth, early metastasis, and a dismal prognosis.[16] The standard of care for first-line treatment is a combination of platinum-based chemotherapy (cisplatin or carboplatin) with etoposide, plus the addition of an immune checkpoint inhibitor, either atezolizumab or durvalumab, followed by immunotherapy maintenance.[18] While this regimen has modestly improved survival compared to chemotherapy alone, nearly all patients will experience disease progression.[16]
Upon relapse, treatment options are limited, and outcomes are poor. The therapeutic strategy is largely dictated by the chemotherapy-free interval (also known as the platinum-free interval), which is the time from the end of first-line platinum therapy to disease progression.[18]
Given the limited efficacy and considerable toxicity of existing second-line therapies, especially for the platinum-resistant population, there remains a profound and urgent unmet medical need for novel, more effective, and better-tolerated treatments.[10]
To address this unmet need, the developers initiated trial NCT05728619, a Phase 1b/2 study designed to evaluate the safety and efficacy of HTMC0435 in combination with temozolomide in patients with relapsed ES-SCLC.[12]
Table 3: Baseline Demographics and Disease Characteristics of Patients at RP2D in Trial NCT05728619 (n=55)
Characteristic | Value | Source(s) |
---|---|---|
Total Patients at RP2D | 55 | 23 |
Brain Metastases | 41.8% (23/55) | 10 |
Liver Metastases | 23.6% (13/55) | 10 |
Prior 1L Platinum + Anti-PD-(L)1 Therapy | 61.8% (34/55) | 10 |
Platinum-Resistant (PFI < 90 days) | 23 patients | 11 |
Platinum-Sensitive (PFI ≥ 90 days) | 29 patients | 11 |
[The high prevalence of brain metastases and the fact that a majority of patients had already received and failed the modern standard of first-line chemoimmunotherapy highlight the advanced nature of the disease in this study population.]
The efficacy results from the trial, based on 49 evaluable patients with a median follow-up of 10.3 months, were presented at the 2025 ASCO Annual Meeting and demonstrated promising antitumor activity.[5]
[A deeper analysis of the efficacy data across predefined patient subgroups reveals critical details about the activity of the HTMC0435 and temozolomide combination. These analyses highlight specific populations that may derive the most significant benefit and provide insights into the drug's mechanism of action.]
[The most striking and clinically significant finding from the trial emerged from the analysis based on prior platinum sensitivity. As previously noted, platinum-resistant disease carries a far worse prognosis than platinum-sensitive disease. Counter to this expectation, the HTMC0435 combination demonstrated robust and comparable efficacy in both populations.]
[The observation of a 12.6-month median OS in a platinum-resistant population is a profound result that significantly exceeds the outcomes typically seen with standard-of-care agents. This suggests that the synthetic lethal mechanism induced by the HTMC0435-TMZ combination is capable of overcoming the biological drivers of platinum resistance. This finding positions the regimen as a potential paradigm-shifting therapy, particularly for the patient population with the highest unmet need.]
[Additional subgroup analyses provided further context:]
[These key efficacy outcomes are summarized and compared in Table 4.]
Table 4: Key Efficacy Outcomes from Trial NCT05728619 in ES-SCLC (Overall and by Subgroup)
Efficacy Endpoint | Overall Population (n=49) | Platinum-Sensitive (n=29) | Platinum-Resistant (n=23) | No Brain Metastases (n=26) |
---|---|---|---|---|
ORR (%) | 24.5% | 26.9% | 25.0% | 38.5% |
DCR (%) | 63.3% | 73.1% | 55.0% | 61.5% |
Median DOR (months) | 6.9 | 4.2 | Not Reached | 5.5 |
Median PFS (months) | 2.8 | 4.2 | 2.5 | 2.7 |
Median OS (months) | 12.0 | 11.2 | 12.6 | 12.0 |
Source(s): [10]
The safety profile of the combination was evaluated in all 59 enrolled patients. While treatment-related adverse events (TRAEs) were common, the overall profile was deemed manageable by the investigators.[10]
Table 5: Summary of Grade ≥3 Treatment-Related Adverse Events (TRAEs) in Trial NCT05728619 (n=59)
Adverse Event | Incidence (%) | Source(s) |
---|---|---|
Any Grade 3-4 TRAE | 55.9% | 11 |
Neutropenia | 35.6% | 10 |
Leukopenia | 28.8% | 10 |
Thrombocytopenia | 13.6% | 10 |
Anemia | 8.5% | 10 |
Dose Reductions due to TRAEs | 10.2% | 11 |
Discontinuations due to TRAEs | 0% | 11 |
The clinical manageability of these side effects is a critical aspect of the regimen's therapeutic potential. Despite the high rate of Grade 3-4 events, dose reductions of HTMC0435 were required in only 10.2% of patients.[11][ Most significantly,]
no patients had to permanently discontinue HTMC0435 due to a TRAE.[11][ This remarkable statistic indicates that with appropriate supportive care (e.g., growth factor support for neutropenia) and potential dose interruptions, clinicians were able to manage the toxicities effectively, allowing patients to remain on therapy long enough to derive a clinical benefit. This manageable safety profile strongly supports a favorable risk/benefit assessment for the combination in this advanced disease setting.]
The investigation of HTMC0435 in advanced biliary tract cancers (BTCs), which include cholangiocarcinoma and gallbladder cancer, is a strategic expansion based on the growing understanding of the molecular drivers of these diseases. A subset of BTCs, particularly intrahepatic cholangiocarcinomas, are known to harbor genomic alterations in DDR pathway genes, including BRCA1/2, ATM, BAP1[, and others. This creates a biological rationale for the use of PARP inhibitors based on the principle of synthetic lethality.]
The current standard of care for first-line advanced BTC is combination chemotherapy (gemcitabine and cisplatin) with the addition of an immune checkpoint inhibitor (durvalumab or pembrolizumab).[24][ For patients who progress, second-line options are limited, especially for those whose tumors do not have specific actionable mutations like]
FGFR2 fusions or IDH1 mutations.[26][ This creates a significant unmet need for effective subsequent-line therapies.]
To explore the potential of HTMC0435 in this setting, a Phase 2 clinical trial (CTR20230615) has been initiated and is actively recruiting patients in China. This trial is designed to evaluate the efficacy and safety of HTMC0435 as a monotherapy in patients with advanced BTC.[7][ Success in this trial could position HTMC0435 as a first-in-class PARP inhibitor for a biomarker-selected population of BTC patients.]
The therapeutic rationale for PARP inhibitors in pancreatic cancer is well-established and clinically validated. Approximately 5-7% of patients with pancreatic adenocarcinoma have a germline mutation in BRCA1 or BRCA2, which confers sensitivity to PARP inhibition.[28][ Based on this, the PARP inhibitor olaparib is approved by the FDA as a maintenance therapy for patients with germline]
BRCA-mutated metastatic pancreatic cancer whose disease has not progressed after at least 16 weeks of a first-line platinum-based chemotherapy regimen.[28]
This precedent provides a clear development path and a defined, biomarker-selected patient population for HTMC0435. The company is actively pursuing this indication with a Phase 2 clinical trial (CTR20230616), which is recruiting patients in China to evaluate HTMC0435 in advanced pancreatic cancer.[7][ This trial will likely aim to demonstrate activity that is competitive with or superior to existing PARP inhibitors in this space, or potentially explore its utility in a broader population of patients with other DDR alterations beyond just germline]
BRCA[ mutations.]
Reflecting a broader strategic vision, the development program for HTMC0435 includes investigations in a wider range of solid tumors. An early Phase 1/2 trial (CTR20200573) was initiated in 2020 to evaluate HTMC0435 monotherapy in patients with various advanced malignant solid tumors, likely to establish its safety and identify signals of activity across different histologies.[29]
More recently, building on the compelling data from the SCLC cohort, a dedicated Phase 2 trial (CTR20233204) was launched to evaluate the HTMC0435 plus temozolomide combination in patients with advanced solid tumors.[7][ This suggests a potential "tumor-agnostic" or "pan-tumor" strategy, where the therapeutic rationale is based on the powerful synergistic mechanism of the combination rather than a specific cancer type. Such a strategy could target tumors known to have high levels of replicative stress or underlying DDR defects, regardless of their tissue of origin.]
[The clinical data for the HTMC0435 and temozolomide combination in relapsed/refractory ES-SCLC positions it as a highly competitive and potentially superior therapeutic option compared to the current standards of care. A direct comparison of survival outcomes highlights its potential to shift the treatment paradigm.]
The median OS of 12.0 months observed in the overall second-line population of the NCT05728619 trial compares favorably to historical data for other approved agents. For instance, topotecan, a long-standing option, is associated with a median OS of approximately 6 months.[11] Lurbinectedin, a more recently approved agent, demonstrated a median OS of 9.3 months in its pivotal trial, primarily in a platinum-sensitive population.[20]
The most compelling competitive advantage for HTMC0435 lies in the platinum-resistant setting. The median OS of 12.6 months achieved by the combination in this population is substantially longer than what is typically observed with other therapies. A recent real-world study of second-line treatments in France reported a median OS of just 5.0 months for platinum-resistant patients.[18][ This stark difference underscores the potential for the HTMC0435 regimen to become the new standard of care for this group, which currently has the most limited options and worst prognosis. A comparative summary is provided in Table 6.]
Table 6: Comparative Efficacy of Second-Line Therapies for ES-SCLC
Therapy | Patient Population | ORR (%) | Median OS (months) | Source(s) |
---|---|---|---|---|
HTMC0435 + Temozolomide | Platinum-Resistant | 25.0% | 12.6 | 10 |
HTMC0435 + Temozolomide | Platinum-Sensitive | 26.9% | 11.2 | 10 |
Lurbinectedin | Platinum-Sensitive | 35.2% | 9.3 | 20 (literature value) |
Topotecan | Platinum-Resistant | ~5-10% | ~5-6 | 11 (literature value) |
Platinum Rechallenge | Platinum-Sensitive | ~20-30% | ~8.1 | 18 |
[In indications beyond SCLC, the strategy for HTMC0435 will likely rely on a biomarker-driven approach to identify patient populations most likely to benefit.]
[Despite the highly promising results, several critical questions must be addressed for HTMC0435 to advance toward regulatory approval and broad clinical adoption.]
A strong intellectual property (IP) portfolio is fundamental to the commercial viability of any new pharmaceutical agent. The available information indicates that there are "100 Patents (Medical) associated with HTMC-0435," although specific details of this patent estate are not publicly accessible.[7][ A comprehensive patent portfolio would be expected to include composition of matter patents, which provide the broadest and most durable protection, as well as patents covering specific methods of use (e.g., for treating SCLC), manufacturing processes, and formulations.]
[The combination of its status as a New Molecular Entity and its Orphan Drug Designation in the United States provides HTMC0435 with a strong foundation for market exclusivity. Upon potential approval, it would benefit from both standard NME data exclusivity and an additional seven years of orphan drug exclusivity for its designated indication, preventing generic competition and providing a protected period to recoup R&D investment. The precise expiration dates of the core composition of matter patents will be a key determinant of the drug's long-term commercial value and a central focus of any due diligence or partnership evaluation.]
[HTMC0435 has emerged as a highly promising investigational PARP1/2 inhibitor with a compelling clinical profile. Its development is founded on the validated and powerful mechanism of synthetic lethality, which has been successfully leveraged by other agents in its class. The current data package, led by the results from the NCT05728619 trial, strongly supports its continued development as a significant new therapy in oncology.]
[The combination of HTMC0435 with temozolomide has produced clinically meaningful and durable responses in a heavily pre-treated population of patients with relapsed/refractory extensive-stage small cell lung cancer, an area of profound unmet medical need. The achievement of a 12.0-month median overall survival in this setting is a standout result. The most significant and potentially practice-changing finding is the regimen's robust efficacy in platinum-resistant patients, a group with dismal outcomes on current therapies. The 12.6-month median OS in this subgroup suggests that HTMC0435 could become a new standard of care.]
[Furthermore, the safety profile, though characterized by significant but expected hematologic toxicities, appears to be highly manageable in the clinical setting, as demonstrated by the remarkable zero percent discontinuation rate due to adverse events. This favorable therapeutic index enhances its potential for broad clinical adoption. The strategic expansion of the clinical program into other DDR-vulnerable tumors, such as pancreatic and biliary tract cancers, indicates a well-considered plan to maximize the asset's therapeutic reach.]
[While the development has thus far been centered in China, the clear signals of a global strategy—including data presentation at ASCO and a U.S. Orphan Drug Designation—are encouraging. The critical next step for HTMC0435 will be to validate its promising Phase 2 results in a large, randomized Phase 3 trial. Successful execution of such a trial, coupled with a coherent global regulatory and commercialization strategy, could establish HTMC0435 as an important new tool for oncologists and a significant therapeutic advance for patients with several difficult-to-treat cancers.]
A. Summary of Key Clinical Trials for HTMC0435
Trial Identifier | Indication | Phase | Status (as of late 2023/early 2024) | Country/Location | Key Intervention |
---|---|---|---|---|---|
NCT05728619 | Extensive-Stage Small Cell Lung Cancer (ES-SCLC) | 1/2 | Active, Recruiting | China | HTMC0435 + Temozolomide |
CTR20230615 | Advanced Biliary Tract Cancer | 2 | Recruiting | China | HTMC0435 Monotherapy |
CTR20230616 | Advanced Pancreatic Cancer | 2 | Recruiting | China | HTMC0435 Monotherapy |
CTR20233204 | Advanced Solid Tumors | 2 | Recruiting | China | HTMC0435 + Temozolomide |
CTR20200573 | Advanced Malignant Solid Tumors | 1/2 | Initiated (May 2020) | China | HTMC0435 Monotherapy |
B. Physicochemical Properties of HTMC0435
Property | Value |
---|---|
Chemical Name | 4-(3-(4-(1H-imidazo[4,5-b]pyridin-5-yl)piperazine-1-carbonyl)-4-fluorobenzyl)phthalazin-1(2H)-one |
CAS Number | 2024616-54-2 |
Molecular Formula | C26H22FN7O2 |
Molecular Weight | 483.51 |
Appearance | Solid |
Solubility | 10 mM in DMSO |
Note: Detailed pharmacokinetic data (e.g., half-life, Cmax, AUC) from human studies are not yet publicly available but were an objective of the Phase 1b portion of trial NCT05728619. |
Published at: September 8, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.