Abstract
Background NUC-3373 is a novel thymidylate synthase (TS) inhibitor designed to directly deliver the active anti-cancer metabolite fluorodeoxyuridine-monophosphate (FUDR-MP or FdUMP) intracellularly. In addition to being a potent TS inhibitor, NUC-3373 has also been shown to cause DNA damage and promote the release of damage-associated molecular patterns. These diverse mechanisms position NUC-3373 as a potentially effective combination partner for several other anti-cancer agents.
Methods NuTide:303 is a Phase Ib/II open label, multi-arm, parallel cohort dose-finding and expansion study designed to evaluate optimal combination partners for NUC-3373. Module 1 planned to enroll up to 12 evaluable patients with advanced/metastatic solid tumors to determine the recommended dose for NUC-3373 in combination with leucovorin (LV) and pembrolizumab. Module 2 planned to enroll approximately 6-12 evaluable patients with advanced/metastatic non-small cell lung cancer (of any histology) or pleural mesothelioma to determine the recommended dose for NUC-3373 in combination with LV and docetaxel. A 3+3 dose escalation design was used in both modules, with safety parameters continually assessed and tumor assessments conducted at screening and every 8 weeks from C1D1 until progression.
Results Thirteen patients were enrolled in Module 1, with 9 evaluable for response. No DLTs were observed in these heavily pre-treated PD-(L)1 experienced patients with a variety of different tumor types. Four patients remained on treatment for >3 months and 3 patients were ongoing at the time of data cut-off. An objective response rate of 22% and a disease control rate of 67% were reported. NUC-3373 + LV + pembrolizumab was well tolerated, the most common treatment-related adverse events (AEs) were nausea, vomiting, and diarrhea and the majority of events were Grade 1 (100%) or 2 (85%). Four patients were enrolled in Module 2, with 3 evaluable for response. Two patients treated at the first dose level achieved stable disease and completed >6 months of treatment The most common treatment-related AEs were fatigue (n=4) and diarrhea (n=3) and the majority of events were Grade 1 (100%) or 2 (100%).
Conclusions Preliminary results from Module 1 show signals of anti-cancer activity and tolerability, providing support for further exploration of this combination in the Phase II part of the study. Preliminary results from Module 2 suggest that the probability of being able to escalate to typical efficacious doses of docetaxel used in combination is low due to overlapping toxicity. Further modifications to include the use of a different taxane in this combination are currently being considered.
Introduction
NUC-3373 is a novel thymidylate synthase (TS) inhibitor that was specifically designed to overcome the key limitations and pharmacologic challenges associated with fluoropyrimidines and other TS inhibitors [1, 2]. TS is a crucial enzyme in the de novo synthesis of deoxythymidine monophosphate (dTMP), a nucleotide essential for DNA synthesis and repair [3]. TS is often upregulated in rapidly dividing cancer cells, making it a critical target for anti-cancer agents. There are several widely used anti-cancer agents that inhibit TS, including 5-fluorouracil (5-FU), capecitabine and pemetrexed; however, their clinical utility is limited by off-target toxicity, inefficient activation pathways, and poor pharmacokinetic (PK) profiles [4-7].
NUC-3373 was designed to directly deliver the active anti-cancer metabolite fluorodeoxyuridine-monophosphate (FUDR-MP or FdUMP) intracellularly, bypassing the complex and inefficient activation pathway that is required for other fluoropyrimidines. NUC-3373 is also resistant to enzymatic breakdown by dihydropyrimidine dehydrogenase (DPD), reducing the generation of the toxic by-product α-fluoro-β-alanine (FBAL) and offering a potential treatment option to patients with DPD deficiencies. We have shown that in addition to being a potent TS inhibitor, NUC-3373 is an effective DNA-damaging agent [8] and promotes release of damage-associated molecular patterns (DAMPs) [9, 10]. These diverse mechanisms position NUC-3373 as a potentially effective combination partner for several other anti-cancer agents, including checkpoint inhibitors.
NuTide:303 is a Phase Ib/II open label, multi-arm, parallel cohort dose-finding and expansion study designed to evaluate optimal combination partners for NUC-3373 in patients with advanced solid tumors [11]. Two modules had been opened as of the data cut-off. Module 1 was designed to investigate NUC-3373 in combination with leucovorin (LV) and pembrolizumab for the treatment of patients with advanced solid tumors. The rationale for this combination is based on data from nonclinical studies showing that NUC-3373 may have the potential to modulate the tumor microenvironment (TME) and prime an anti-tumor immune response. In colorectal cancer (CRC) cell lines, NUC-3373 was shown to cause DNA damage and promote the release of DAMPs, including high-mobility-group box 1 (HMGB1), calreticulin and ATP [9, 10]. DAMPs are known to enhance cancer cell recognition by the immune system, promoting recognition and destruction by inflammatory and immune mechanisms. In co-culture experiments, NUC-3373 treatment caused an increase in the surface expression of PD-L1 on CRC and non-small cell lung cancer (NSCLC) cells, indicating that NUC-3373 may enhance the anti-cancer potential of immune checkpoint inhibitors targeting the PD-1/PD L1 axis [12, 13]. In support of this, CRC and NSCLC cells pre-treated with NUC-3373 and co-cultured with peripheral blood mononuclear cells (PBMCs) displayed a reduction in cell viability compared to monocultured CRC or NSCLC cells. Furthermore, NUC-3373 was shown to enhance cell death in CRC and NSCLC cells co-cultured with PBMCs, with NUC-3373 potentiating the activity of the PD-1 inhibitors nivolumab (in CRC cells) and pembrolizumab (in NSCLC cells) [12, 13]. Thus, NUC-3373 is an attractive combination partner for PD-(L)1 inhibitors.
Module 2 was designed to investigate NUC-3373 in combination with LV and docetaxel for the treatment of patients with NSCLC or pleural mesothelioma. The rationale for this module is based on data from nonclinical studies showing that NUC-3373 is a more potent TS inhibitor than either 5-FU or pemetrexed in NSCLC cells. In a set of in vitro studies in four human NSCLC cell lines, NUC-3373 showed cytotoxicity across all cell lines with half-maximal inhibitory concentration (IC50) values in the low µM range. Consistent with existing literature, the squamous cell carcinoma cells lines (Cx140 and Nx002) had higher TS expression levels than the adenocarcinoma cell lines (A549 and Calu3) [14]. However, treatment with NUC-3373 promoted TS inhibition in all four cell lines, showing that NUC-3373 is able to inhibit TS regardless of basal TS expression levels and cell histology [14]. These results suggest that NUC-3373 may have activity where pemetrexed has previously failed in lung cancers with squamous cell differentiation.
The nonclinical findings have been substantiated in patients treated with NUC-3373 as a monotherapy and in combination with other agents in clinical studies [15-19]. NUC-3373 has been well-tolerated and has shown a favorable PK profile.
These data demonstrate the potential for NUC-3373 to offer an effective treatment option and combination partner in several tumor indications. Here we describe initial results from the Phase Ib part of Modules 1 and 2 of the NuTide:303 clinical study.
Methods
Study Design and Objectives
NuTide:303 is a Phase Ib/II open label, multi-arm, parallel cohort dose-finding and expansion study designed to evaluate the safety, efficacy and PK of NUC-3373 in combination with other agents for the treatment of patients with different tumor types (EudraCT 2022-000722-14; NCT05714553). This study is modular, with each module evaluating a different NUC-3373 combination, and consists of a dose-validation phase (Phase Ib) and a potential dose-expansion phase (Phase II). Approximately 6-20 evaluable patients were planned to be enrolled in the Phase Ib part of each module to determine safety, tolerability, and preliminary efficacy of NUC-3373 in combination with other agents. Following review of the Phase Ib data, each module may then move into Phase II to enable a further assessment of safety and efficacy in approximately 20-40 patients.
In Module 1, up to 12 evaluable patients were planned to be enrolled in Phase Ib to determine the recommended dose for NUC-3373 + LV in combination with pembrolizumab. In Phase II, a further evaluation of the combination may be performed in exploratory, signal-seeking cohorts of approximately 20-40 patients in 2-4 selected tumor types that have shown encouraging signs of activity (clinical or translational) during Phase Ib.
In Module 2, approximately 6-12 evaluable patients were planned to be enrolled in Phase Ib to determine the recommended dose for NUC-3373 + LV in combination with docetaxel. In Phase II, a further evaluation of the combination may be performed using a Simon’s two-stage design [20].
The study was conducted at 3 clinical sites in the United Kingdom and was performed in accordance with the Declaration of Helsinki and the principles of Good Clinical Practice. The protocol was approved by the relevant Ethics Committees and all patients provided written informed consent before undergoing any study procedures.
Patients
Module 1
Patients with advanced/metastatic solid tumors who had progressed on ≤2 prior therapies for metastatic disease, that may have included 1 prior immunotherapy-containing regimen (either monotherapy or in combination with chemotherapy) or who had not progressed but where addition of NUC-3373 + LV to standard pembrolizumab monotherapy may have been appropriate (i.e., patients who had exhausted all other treatment options for advanced disease) were eligible. Patients must have had tumor types for which pembrolizumab is approved and is a suitable treatment option (e.g., melanoma, NSCLC, urothelial carcinoma, oesophageal carcinoma, renal cell carcinoma, head and neck squamous cell carcinoma, cutaneous squamous cell carcinoma, MSI high CRC, gastric cancer, triple negative breast cancer, classical Hodgkin lymphoma, and endometrial carcinoma). Patients must also have had radiologically measurable disease, Eastern Cooperative Oncology Group (ECOG) performance status 0-1, and adequate haematologic, renal and hepatic function. Patients with known neutralizing antibodies against checkpoint inhibitors or who had discontinued prior therapy due to toxicity attributed to checkpoint inhibitors were excluded.
Module 2
Patients with advanced/metastatic NSCLC (of any histology) or pleural mesothelioma who had progressed on, or were unable to tolerate, 1 or 2 prior lines of cytotoxic chemotherapy-containing regimens for advanced/metastatic disease (not including neoadjuvant or adjuvant therapy). Additional prior lines of treatment with targeted agents or immunotherapy were allowed as long as they were not given in combination with cytotoxic chemotherapy. Patients must also have had radiologically measurable disease, ECOG performance status 0-1, and adequate hematologic, renal and hepatic function. Patients who had received prior treatment with docetaxel for advanced/metastatic disease were excluded.
Treatment
Module 1
An initial cohort of 3 patients were to receive NUC-3373 at a dose of 1875 mg/m2 plus LV (400 mg/m2) on Days 1, 8 and 15 in combination with pembrolizumab (200 mg) on Day 1 of 21-day cycles. If this dose was tolerated (i.e., no DLTs in the first 3 patients), a further 3 patients were to be enrolled at the same dose level. Following this, the cohort could have been expanded up to a total of 12 patients.
Module 2
An initial cohort of 3 evaluable patients were to receive NUC-3373 at a dose of 750 mg/m2 plus LV (400 mg/m2) on Days 1 and 22 and docetaxel (55 mg/m2) on Day 8 of 28-day cycles. If this dose was tolerated, dose escalation was to proceed in a modified 3+3 design. The dose and schedule for docetaxel was slightly modified from the standard regimen used in advanced
NSCLC, in which docetaxel is typically administered every 3 weeks at a dose of 75 mg/m2, to a 4-weekly administration schedule. This modification was for safety reasons to avoid concomitant treatment with a new agent (NUC-3373) during the post-docetaxel white blood cell nadir. Following treatment of 4 patients at the first dose level, dosing was stopped due to toxicity challenges with docetaxel, which required dose reductions to 35 mg/m2 in 1 patient from Cycle 3 onwards.
Clinical Assessment
All patients who received at least one dose of study treatment were included in the safety analysis population. Safety parameters were continually assessed. Treatment-emergent adverse events (TEAEs) were graded according to the National Cancer Institute Common Terminology Criteria for Adverse Events. Patients were defined as evaluable for DLT assessment if they had received at least 75% of the intended dose of each agent during Cycle 1 (Module 1) or Cycles 1 and 2 (Module 2) or experienced a DLT. The definition of DLT is provided in the Supplementary Data.
Tumor response assessments were performed for the evaluable population defined as patients with measurable disease at baseline who received at least two cycles of treatment, received at least 75% of planned treatment over the two cycles, and had a post-treatment objective disease assessment. Computed tomography (CT)/magnetic resonance imaging (MRI) based tumor assessments were conducted at screening and every 8 weeks from C1D1 until progression. Responses from all available post-baseline scans from evaluable patients were used for derivation of the best overall response (BOR) and disease control rate (DCR). A response of stable disease was only considered if the scan was taken at least 7 weeks after the baseline scan. The highest reduction of the sum of diameters of target lesions was selected and reported for each patient with available post-baseline scan. This was expressed as a percentage change from baseline. In Module 1, the primary efficacy endpoint (ORR) was assessed using iRECIST criteria and the secondary efficacy response endpoints were assessed using RECIST v1.1 criteria. In Module 2, all efficacy response endpoints were assessed using RECIST v1.1 criteria. Responses (CRs and PRs) had to be confirmed by repeated measurement at least 28 days and not more than 42 days after initial documentation. As per iRECIST, assessment of disease progression also required confirmation of PD in Module 1. Patients were to be treated and followed-up post-initial progression (termed unconfirmed PD or iUPD) until subsequent disease progression (termed confirmed PD or iCPD) was confirmed.
Statistical Analyses
Since this is an exploratory study focused on tolerability, translational data, and signal-seeking activities, descriptive statistics were used.
Module 1
In Phase Ib, up to approximately 12 evaluable patients in total were to be enrolled to determine the tolerability of NUC-3373 in combination with pembrolizumab.
Module 2
In Phase Ib, approximately 6-12 evaluable patients were to be enrolled in a 3+3 dose escalation design to determine the recommended dose for NUC-3373 in combination with docetaxel.
The data presented have a cut-off date of 08 October 2024.
Results
Patient Characteristics
Between March 2023 and July 2024, 17 patients were enrolled across 3 centers in the UK (Table 1).
A total of 13 patients were enrolled in Module 1, one was not evaluable due to a pre-existing cardiac arrythmia. All patients had metastatic disease and the primary tumor types were cutaneous melanoma (n=3), urothelial carcinoma of bladder (n=2), oropharyngeal (n=2), NSCLC (n=2), anal (n=1), sinonasal (n=1), pancreatic (n=1), and pleural mesothelioma (n=1). Patients had received a median of 1 prior line of therapy (range: 0-2) for metastatic disease. Nine patients had received prior immunotherapy treatment, 6 of whom (67%) had disease progression on treatment.
A total of 4 patients were enrolled in Module 2. All patients had metastatic disease and the primary tumor types were NSCLC (n=3) and pleural mesothelioma (n=1). Patients had received a median of 2 prior lines of therapy (range: 1-3) for metastatic disease, and one patient had received prior treatment with paclitaxel.
Further details for individual patients are provided in the Supplementary Data.
Treatment Exposure
Module 1
The DLT evaluation period was 1 cycle in Module 1. No DLTs were observed following treatment of the first 6 patients with 1875 mg/m2 NUC-3373 + 400 mg/m2 LV + 200 mg pembrolizumab; therefore, the cohort was expanded to 12 patients at the same doses to confirm the recommended dose. Time on treatment was encouraging in this group of heavily pre-treated PD-(L)-1 experienced patients with a variety of different tumor types, with 3 patients ongoing at the time of data cut-off (Figure 1).
Module 2
The DLT evaluation period was 2 cycles in Module 2. No DLTs were observed following treatment of the first 4 patients with 750 mg/m2 NUC-3373 + 400 mg/m2 LV + 55 mg/m2 docetaxel; however, overlapping toxicities of diarrhea, nausea, vomiting, mucositis and fatigue were observed and enrolment to this module was put on hold. Despite this, two patients treated at the first dose level completed more than 6 months of treatment (Figure 2).
Following a Safety Review Committee meeting, the decision was made to stop further enrolment in this module due to tolerability challenges associated with docetaxel. Although no DLTs were observed at this first dose level, the toxicity (largely attributed to docetaxel) was such that it was felt that further dose escalation to single-agent efficacious dose levels of either agent would be difficult. However, given the signs of efficacy of the NUC-3373 + taxane combination, the decision was made to explore other taxanes which may be easier to combine with NUC-3373, rather than proceeding with the NUC-3373 + docetaxel combination.
Safety
Module 1
Thirteen patients were included in the safety population in Module 1. The most common treatment-related TEAEs were nausea (69%), vomiting (69%), diarrhea (46%), fatigue (38%), AST increased (31%), and infusion-related reaction (31%). The vast majority of treatment-related TEAEs were Grade 1 (100%) or 2 (85%), only one patient had a Grade 3 TEAE of hyponatremia (Table 2). No Grade 4 or 5 treatment-related TEAEs were reported.
Module 2
Four patients were included in the safety population in Module 2. The most common treatment-related TEAEs were fatigue (n=4), diarrhea (n=3), hypotension (n=2), and stomatitis (n=2). The majority of treatment-related TEAEs were Grade 1 (100%) or 2 (100%), with four Grade 3 TEAEs of fatigue, diarrhea, hypotension, and vomiting reported (Table 3). No Grade 4 or 5 treatment-related TEAEs were reported.
This module was put on hold due to tolerability challenges with docetaxel at the lowest dose level, as described in the treatment exposure section.
Efficacy
Module 1
Nine patients were evaluable for response. Signals of anti-cancer activity were observed, with tumor shrinkages in 2 patients (Figure 3).
The best overall response was iPR for 2 patients, iSD for 4 patients, unconfirmed iPD for 2 patients and confirmed iPD for 1 patient, resulting in an objective response rate (ORR) of 22% and a disease control rate (DCR) of 67%. One patient had a 100% reduction in the sum of target lesions, but non-target lesions were present and so was not considered a complete response (iCR). Another patient had a confirmed partial response (iPR) with an 81% reduction in tumor volume.
Further information on the two patients with PRs is presented in the case studies below.
A patient with urothelial carcinoma of bladder (Lynch syndrome) had received 2 prior lines of therapy. They received gemcitabine plus cisplatin in the adjuvant setting (discontinued after 2 months due to myelosuppression) and atezolizumab in the metastatic setting (best response of PR and discontinued after 23 months as had received maximum allowable therapy). They had 1 target lesion in the lung. The patient received treatment with 1875 mg/m2 NUC-3373 plus 200 mg pembrolizumab and had a 100% reduction in the target lesion; however, non-target lesions were present so this response was considered a confirmed iPR and not a iCR. The patient remains ongoing on treatment after 10 months and has not had any dose reductions.
A patient with cutaneous melanoma had received 2 prior lines of therapy. They received pembrolizumab (best response of PD within 5 months) and trametinib plus dabrafenib (discontinued trametinib after 1 month due to toxicity and achieved best response of SD on dabrafenib before experiencing PD after 7 years). They had 1 target lesion in the lymph nodes (bilateral). The patient received treatment with 1875 mg/m2 NUC-3373 plus 200 mg pembrolizumab and had an 81% reduction in tumor volume (confirmed iPR). The patient remains ongoing on treatment after 12 months and has not had any dose reductions.
Module 2
Three patients were evaluable for response. The best overall response was SD for 2 patients and PD for 1 patient. Further information on the two patients with SD is presented in the case studies below.
A patient with pleural mesothelioma had received 3 prior lines of therapy. They received cisplatin plus pemetrexed (PD within 4 months), nivolumab (PD within 4 months), and carboplatin plus pemetrexed (PD within 1 month). This patient had progressive disease and a high tumor burden at study entry. They had 4 target lesions (2x lymph node and 2x mediastinum). The patient received treatment with 750 mg/m2 NUC-3373 plus 55 mg/m2 docetaxel and achieved prolonged SD of more than 13 months. The patient received 4 cycles of NUC-3373 plus docetaxel followed by 5 cycles of NUC-3373 alone before coming off treatment due to patient choice (treatment fatigue). The patient has not experienced PD and the Investigator believes the patient derived clinical benefit (durable disease control) from NUC-3373.
A patient with NSCLC (squamous) had received 2 prior lines of therapy. They received carboplatin plus paclitaxel plus pembrolizumab (SD for 2 months) and pembrolizumab as maintenance (PD within 21 months). They had 1 target lesion in the lung. The patient received treatment with 750 mg/m2 NUC-3373 plus 55 mg/m2 docetaxel and achieved prolonged SD of 7 months. The patient received 6 cycles of NUC-3373 plus docetaxel followed by 2 cycles of NUC-3373 alone before coming off treatment due to disease progression.
Discussion
This Phase Ib/II modular study was designed to evaluate optimal combination partners for NUC-3373 in patients with advanced solid tumors. Two modules have been opened to date and here we present initial data from the Phase Ib part of Modules 1 and 2. In Module 1, NUC-3373 was evaluated in combination with LV and pembrolizumab for the treatment of patients with advanced solid tumors. In Module 2, NUC-3373 was evaluated in combination with LV and docetaxel for the treatment of patients with NSCLC or pleural mesothelioma.
Preliminary results from Module 1 show signals of anti-cancer activity, including meaningful clinical benefit in some patients, and tolerability, providing support for further exploration of NUC-3373 plus pembrolizumab in the Phase II part of the study. These early clinical results build on nonclinical studies and provide further support that NUC-3373 may promote an anti-tumor immune response and potentiate the activity of immune checkpoint inhibitors. In previous in vitro studies it has been shown that NUC-3373 binds to TS which accumulates in the cytoplasm and leads to release of DAMPs [9, 10], which were predicted to increase the likelihood of an anti-tumor inflammatory response.
Preliminary results from Module 2 suggest that, although no DLTs were observed at the first dose level, the toxicity (largely attributed to docetaxel) was such that further dose escalation to single-agent efficacious dose levels of either agent would have been difficult. Given that the docetaxel starting dose (55 mg/m2) was lower than the standard dose used in NSCLC (75 mg/m2), dose de-escalation of docetaxel was not considered a viable option. However, based on signs of efficacy observed with the NUC-3373 + taxane combination, protocol modifications allowing substitution of a different taxane are being explored in an effort to develop a more tolerable regime. Docetaxel is the current standard of care for NSCLC patients without targetable alterations who progress on PD-(L)1 inhibitor-based therapy; however, it is associated with modest clinical benefit (median progression-free survival of 3-4 months) and substantial toxicity [21]. Achieving disease stabilization in hard-to-treat populations such as NSCLC and mesothelioma further supports the hypothesis that NUC-3373 may be an attractive treatment option for primary lung malignancies based on nonclinical studies showing that NUC-3373 is a potent TS inhibitor.
Overall, initial data from NuTide:303 suggest that NUC-3373 may be an effective combination partner for pembrolizumab in a variety of solid tumors. Although the current combination of NUC-3373 and docetaxel may not be tolerable, alternative NUC-3373 regimens with a taxane could be a promising treatment option in lung cancer and are under consideration.
Data Availability
All data produced in the present study are contained in the manuscript and supplementary data
Footnotes
Disclosure: GM has performed consulting and advisory roles at Roche, Boehringer Ingelheim, Mina therapeutics, D2G, BMS, MSD, and AstraZeneca; has participated in speakers’ bureaus for Roche, BMS, MSD, AstraZeneca, Servier, and Bayer; and has received research funding from BMS, MSD, AstraZeneca, and Plexxikon. JS owns stock in Epsilogen Ltd (co-founder) and Avacta Ltd; holds board membership at Apobec Discovery Ltd; has performed advisory roles at Apobec Discovery Ltd, Avacta Ltd, AstraZeneca, BioNTech, BMS and Roche; has received assistance with attendance at international meetings from Starpharma and MSD; and has received reimbursement for recruitment to clinical trials from Achilles, Gilead, GSK, IO Biotech, MSD, Roche, RS Oncology, and Starpharma. RW has performed consultancy and advisory roles at Alcimed, Amphista Therapeutics, BMS, Boehringer Ingelheim, CV6 Therapeutics, NuCana, RIN Institute and Takeda; has received assistance with attendance at international meetings from NuCana and Takeda; and has received honoraria for educational meetings from Bayer, Merckgroup, Pierre Fabrier, Servier and Takeda. DJH is a part-time employee of NuCana. EO and JB are full-time employees of NuCana. FT has performed consulting and advisory roles at CytomX, Kite, Gilead, Scenic Biotech, Immatics, T-Knife Therapeutics, F-Star, Grey Wolf Therapeutics, and AstraZeneca.
Funding: This study was funded by NuCana plc.