Abstract
Preclinical models of cancer have demonstrated enhanced efficacy of cell-cycle checkpoint kinase inhibitors when used in combination with genotoxic agents. This combination therapy is predicted to be exquisitely toxic to cells with a deficient G1–S checkpoint or cells with a genetic predisposition leading to intrinsic DNA replication stress, as these cancer cells become fully dependent on the intra-S and G2–M checkpoints for DNA repair and cellular survival. Therefore, abolishing remaining cell-cycle checkpoints after damage leads to increased cell death in a tumor cell–specific fashion. However, the preclinical success of these drug combinations is not consistently replicated in clinical trials. Here, we provide a perspective on the translation of preclinical studies into rationally designed clinical studies. We will discuss successes and failures of current treatment combinations and drug regimens and provide a detailed overview of all clinical trials using ATR, CHK1, or WEE1 inhibitors in combination with genotoxic agents. This highlights the need for revised patient stratification and the use of appropriate pharmacodynamic biomarkers to improve the success rate of clinical trials.
The design of a significant percentage of clinical trials assessing the efficacy of intra-S and G2–M checkpoint kinase inhibitors in combination with standard-of-care chemo- or radiotherapy is not sufficiently informed by the fundamental biological and mechanistic insights obtained in in vitro assays and animal studies. Here, we summarize the emerging preclinical concepts underlying genotoxic cancer treatments that exploit loss of checkpoint function as a result of WEE1, ATR, ATM, or CHK1/2 inhibition. Furthermore, we suggest how these emerging insights could improve the rational design of clinical studies by dictating appropriate drug regimens, stratifying patient populations harboring defined genetic alterations, and identifying suitable biomarkers to monitor response, with the aim of improving the success of combination therapy in clinical trials.
Introduction
The majority of chemotherapeutics, as well as radiotherapy, either directly damage DNA or target basic cellular and metabolic processes that lead to indirect DNA damage. The DNA damage response (DDR) elicits activation of cell-cycle checkpoints to promote repair by pausing the cell cycle or, in cases of unrepairable DNA damage, stimulate programmed cell death (1, 2). As such, much effort has been directed to augment the efficacy of chemotherapeutics and radiotherapy through combination with cell-cycle checkpoint kinase inhibitors as this forces cells to progress through the cell cycle in the presence of unrepairable DNA damage, ultimately leading to cell death (3). Hence, regimens of combined chemotherapy or radiotherapy with checkpoint kinase inhibitors have proven beneficial in several preclinical studies and the biology and mechanisms behind these combinations have been extensively reviewed (4–6). These preclinical observations have led to the assessment of many of these combination therapies in clinical trials in oncology and, as such, this article provides a perspective on the challenges of reliably translating the robust preclinical findings into clinical benefit.
The majority of cancer cells harbor genomic alterations, most commonly (50%–70%) in the form of a dysfunctional G1 checkpoint, due to loss of function of p53, p21, or p-Rb, greatly enhancing their dependency on the remaining checkpoints for the repair of DNA damage (6–9). Therefore, combining chemotherapeutics or radiotherapy with compounds targeting (intact) intra-S and G2–M checkpoint kinases are predicted to have favorable clinical outcomes based on the rationale that tumor cells lacking any functional checkpoints will not be able to arrest the cell cycle and repair DNA damage (reviewed in refs. 3, 6, 10, and 11). This irreversible damage results in replicative catastrophe or is taken through into mitosis, where tumor cells undergo programmed cell death (6, 12). Several druggable targets have been identified that can be inhibited to disrupt the intra- S and G2–M checkpoints. In the presence of replication stress or DNA damage, cyclin-dependent kinase (CDK) activity is restrained by different checkpoint kinases, including ATR and ATM, WEE1, and checkpoint kinases 1 (CHK1) and 2 (CHK2) (reviewed in refs. 13, 14; Fig. 1, Box 1).
In G1, DSBs are sensed by Ku70/80, which activates DNA-PK, ATM, CHK2, and p53, leading to elevated p21 expression, CDK inactivation, and ultimately G1 arrest, thereby allowing for repair of damage by non-homologous end-joining (NHEJ) before DNA replication initiation (13).
Intra-S checkpoint proteins (ATR, CHK1, and WEE1) can regulate DNA replication origin firing by modulating CDK activity (15, 16). Replication stress and DSB end-resection, essential for repair via homologous recombination, generate single-stranded DNA (ssDNA), which serves as a platform for RPA binding and signaling through the ATR–CHK1 axis to protect stalled forks, and induce deoxyribonucleotide (dNTP) production, thereby ensuring complete replication (17, 18).
G2–M checkpoint proteins, including CHK1 and WEE1, regulate CDK1 activity to control mitotic entry prior to completed DNA repair to prevent mitotic catastrophe (19, 20).
Activity of the CDK1–cyclin B complex, which becomes more active through S-phase and reaches peak activity at the G2–M transition (Fig. 2), is directly restricted by phosphorylation events through WEE1/MYT1, which catalyzes inhibitory tyrosine-15 phosphorylation of CDK1/2. ATR and CHK1 can restrict CDK activity by inhibiting the CDC25 family of phosphatases, responsible for dephosphorylation of tyrosine 15 (21). Hence, control over the balance of CDC25-WEE1 regulation is essential for establishing and maintaining both the intra-S and G2–M checkpoints (14).
Targeted cell death by combination therapies, resulting from elevated CDK activity, occurs through two main pathways: replicative and mitotic catastrophe, although they are not necessarily mutually exclusive. First, ATR, CHK1, and WEE1 play an important role in regulating CDK activity within the S-phase checkpoint (18, 22). Inhibition of these checkpoint kinases can lead to higher CDK activity in S-phase and elevated replication origin firing (15), thereby increasing the cellular demand for dNTPs. Moreover, ATR, CHK1, and WEE1 are critical for deoxyribonucleotide triphosphate (dNTP) production in response to replication stress (23) through promoting the synthesis (24), activation, and stabilization (25, 26), of ribonucleotide reductase. Therefore, their inactivation can lead to under-replication, and depending on the mutational background, replicative catastrophe (18, 27–29). Second, regions of under-replicated DNA could be taken through into mitosis, where mitotic catastrophe is induced through chromosome missegregation or delayed cell death after micronucleation (reviewed in ref. 30). Inhibiting the DDR through abrogation of the damage induced G2–M checkpoint, either via CDC25A stabilization by CHK1 or ATR inhibition or via CDK1 re-activation through WEE1 inhibition, poses additional cytotoxicity (11, 31). As cells subjected to G2–M checkpoint inhibition cannot downregulate CDK1 activity, they enter mitosis and attempt to perform chromosome segregation with extensive DNA damage, ultimately leading mitotic catastrophe and cell death (Fig. 2).
Even though some chemotherapeutic agents have the ability to kill cells throughout the cell cycle, targeting all proliferating cells, some commonly used chemotherapeutic agents display cell cycle phase–specific toxicity, exerting a lethal effect that is maximal in a specific stage of the cell cycle (Fig. 3; refs. 32, 33). It is useful to distinguish between agents that generate actual DNA damage and therefore induce the DDR (e.g., radiotherapy, which leads to production of free oxygen radicals in the cell, or chemotherapy drugs, such as etoposide) from those that primarily act by blocking replisome activity. The replisome is the molecular machinery that performs parental strand separation and DNA synthesis and its inhibition therefore increases replication stress and targets S-phase cells specifically. For example, while replisome poisons initially induce stalled replication forks, and therefore activate the ATR–CHK1 axis, these blocks can ultimately result in DNA damage if the fork is not protected or eventually processed into a DNA double-strand break (DSB), thereby inducing other checkpoint kinases, including ATM and CHK2. As such, targeting specific checkpoint kinases requires an in depth understanding of their roles within the DNA damage and replication stress checkpoint response. For example, ATR and WEE1 inhibition exhibit different modes of action (Fig. 1; ref. 34). In contrast to CHK1 and ATR, WEE1 is not only central to the replication stress response, but also functions as a core regulator of CDK activity (14). Mechanistically, it is also important to note that screens identified CDC25A inactivation as the top gene “hit” conferring resistance to ATR and CHK1 inhibitors (35, 36), while the main determinants for WEE1 inhibitor resistance included CDK2, SKP2, and CUL1 inactivation (37), all required for timely entry into, or progression through, S-phase. Furthermore, as ATR functions in the very early stages of the DDR pathway, alternative methods to activate CHK1 do exist (38). This suggests there might be some differential response to ATR, CHK1, and WEE1 inhibitors in the clinic, depending on the genetic background of the patient's cancer. Therefore, selecting appropriate checkpoint inhibitors (ATM, ATR, CHK1/2, or WEE1) that target one stage of the cell cycle in combination with agents that are maximally cytotoxic during that stage of the cell cycle provides the rationale for combination and multiple rounds of treatment.
In this perspective, we identify four key areas for improvement in the translation of preclinical observations of combination therapies into rationally designed clinically trials: (i) Selection of drug combination partners – clinical trials need to take into account the phases of the cell cycle where anticancer drugs exert their lethal and blocking effects as identified in preclinical studies; (ii) Sequence of drug administration – as cells that do not harbor any DNA damage are not dependent on checkpoint kinases for survival, DNA damaging agents have to be administered at an appropriate time prior to checkpoint kinase inhibition, to achieve the desired antitumor effect; (iii) Appropriate dose reductions – preclinical data shows that in the context of combination therapy, less than the MTD of anticancer therapeutics produces therapeutic benefit. Translating this into the clinical setting could reduce levels of toxicity to the normal tissue; and (d) Patient stratification – patient stratification is essential for effective treatment, based on their genetic alterations and biological insights obtained during preclinical and phase I studies.
Here, we provide a perspective on preclinical studies testing “cyclotherapy” (reviewed in ref. 39), defined as a strategy that exploits loss of checkpoint function, through use of inhibitors or due to genetic defects, in combination with DNA-damaging agents. We argue that in combination therapy, the selection of DNA-damaging agents should not be based upon the cancer subtype, but rather on cell-cycle specificity and the genetic background of the tumor. Furthermore, we briefly describe pharmacodynamic biomarkers that can be used to monitor the success of such combination regimens. We hope that this perspective will shed new light on the rational design of cyclotherapy trials.
Preclinical: Sensitivity to Combination Therapies
Choice and scheduling of combination therapies
The combination of intra-S and G2–M checkpoint kinase inhibitors and genotoxic agents has been extensively studied in preclinical models to determine the most effective drug combinations. In Fig. 3, we classified chemotherapy agents based on their action during certain phases of the cell cycle and several preclinical studies have demonstrated the importance of this concept when choosing drug combination partners. For instance, Montano and colleagues showed that SCH9000776 (CHK1i) sensitizes cells to hydroxyurea (20- to 70-fold) and gemcitabine (5- to 10-fold), but not to cisplatin or SN-38 (active metabolite of irinotecan), a topoisomerase I inhibitor, in a number of cell lines (40). SN-38 induces cell-cycle arrest when the replication fork collides with an inhibited topoisomerase I, resulting in DNA DSBs that subsequently activate both Chk1 and Chk2 (41, 42). Even though Chk1 is required for homologous recombination (HR)-mediated repair of these DSBs, ATM-Chk2 is essential to the G2–M arrest, and hence, predominates over the control of cellular survival in this context (41). It is not surprising, therefore, that concurrent incubation of SN-38 with the ATM inhibitor KU55933 caused a dramatic sensitization (40). Similarly, Chk2, but not Chk1, depletion dramatically increased the proportion of apoptotic HCT116 cells exposed to camptothecin (CPT), another topoisomerase I inhibitor (41). Even though the clinical use of CPT was hindered due to severe toxicity and low solubility and stability issues, many other semisynthetic CPT derivatives, including irinotecan and topotecan, continue to be important anticancer agents (43). Therefore, the relevance of cell-cycle phase specific cytotoxicity, and the relative importance of the canonical ATR-Chk1 and ATM-Chk2 pathways in each of those stages, needs to be considered in the design of combination therapies.
Not only the choice of drugs, but also the sequencing and schedule of chemotherapy agents and cell -cycle checkpoint inhibitors impacts on the effectiveness of combination therapy. In many cases, delayed, but not concurrent, treatment with checkpoint inhibitors sensitizes cancer cells to S-phase–specific chemotherapeutic agents (44). For example, the combination of CHK1i with gemcitabine or hydroxyurea (HU) is only effective when CHK1 inhibition occurs >18 hours after treatment with these chemotherapy agents (Fig. 3) as shown by experiments in a number of cell lines, in vivo patient-derived xenografts, and ex vivo studies (40, 45, 46). Gemcitabine and HU are both examples of clinically approved inhibitors of ribonucleotide reductase (RR), which catalyzes the rate-limiting step of the formation of dNTPs, the precursors of DNA synthesis, by direct reduction of the corresponding ribonucleotides (47, 48). Hence, through inhibition of RR, gemcitabine and HU limit synthesis of dNTPs such that replication slows or stops, and therefore cause an accumulation of S-phase–arrested cells only after prolonged treatment, where RPA-coated replication forks become more dependent on Chk1 for stability (40, 46). Therefore, as chemotherapeutic agents cause replication to slow down over increasing time, cells become more sensitive to inhibition of Chk1 (44, 49). Similar results have been found in preclinical experiments using NCI-H2122 and NCI-H441 non–small cell lung cancer cell lines and in vitro/in vivo models with combination therapy of CHK1i (LY2603618) and pemetrexed, which disrupts the production of folate, essential for DNA replication. The activity of LY2603618 manifested only after pemetrexed had caused maximum S-phase arrest, which required prolonged drug exposure (50). Furthermore, cells that had already completed DNA synthesis were not likely to be sensitive to the combination of these agents, and as such, it takes time for an entire asynchronous population of cells to traverse S-phase before an effect was observed, and multiple rounds of treatment might therefore be beneficial (50).
Dosage
Combinations of checkpoint kinase inhibitors and chemotherapy agents will be additively toxic and as such, doses should be reduced appropriately when drugs are used in combination (32, 51). The objective should be to identify a combination schedule that effectively inhibits the target [i.e., the minimum biologically effective dose (BED); ref. 52] rather than the MTD for the checkpoint inhibitor, in combination with the appropriately timed induction of the required DNA damage by the combination partner (cytotoxic or radiation induced). This is particularly important in terms of developing a schedule for combination therapies, where tolerability over time is important to maximize clinical benefit (51). If the drug regimen is designed rationally, and phase-specific agents are added at the cell-cycle phase where they can exert their maximal effect, one may not need to use the MTD of each individual drug to exert an antitumor effect, thereby limiting toxicity-related issues (51). One of the important aspects of the translation of preclinical data into the clinic is the selection of a starting dose for in-human trials (53), which is complex for single-agent trials, but even more so for combination therapies. Detailed analysis and understanding of both human and mouse pharmacokinetics for the agents concerned as well as their interactions are required, and discussion of these complex issues is beyond the scope of this review.
Preclinical studies of CHK1i in combination with HU in MDA-MB-231 breast cancer cells, revealed that the addition of CHK1 inhibitors SCH900776 (1.25 μmol/L) or UCN-01 (200 nmol/L) caused a 60-fold decrease in the IC50 for HU (40), and these results were further validated in a number of other cell lines. Similar reductions in IC50 values were found for gemcitabine and cytarabine, which interferes with DNA synthesis, in MCF10A cells, when these drugs were combined with SCH900776 (CHK1i; ref. 40).
In many cases, continuous exposure (>24 hours) to a checkpoint kinase inhibitor or a chemotherapy agent alone, such as CHK1 inhibitors (MK-8776 or LY2606368), cisplatin or the WEE1 inhibitor (AZD1775) at high concentrations (IC80) is insufficient to robustly induce γH2AX, which provides a read-out of endogenous DNA damage and causes subsequent cytotoxic effects (49, 54, 55). Combination treatments, however, including CHK1 inhibition and cisplatin, CHK1 inhibition and radiation, and third, WEE1 inhibition (AZD1775, also known as adavosertib) and irinotecan, demonstrate greater-than-additive induction of γH2AX in the majority of cells (45%–75%; refs. 55–57) and hence, the concentrations of these agents may be greatly reduced when used together.
Biomarkers and genetic background
As discussed previously, intra-S and G2–M cell-cycle checkpoint inhibitors are especially effective when administered in the context of a dysfunctional G1–S checkpoint, due to the inactivation of endogenous inhibitors (p16 or p21 family) or p53-mediated changes, or alterations causing premature S-phase entry (CCNE1 amplification, RB1 loss, or CDKN2A mRNA downregulation; refs. 5, 58). For example, the WEE1 inhibitor (WEE1i) MK-1775 (AZD1775) was shown to preferentially kill p53-deficient tumors by using p53 matched-pair cell lines (54). As the G1 checkpoint is compromised due to TP53 loss and these cancer cells thus rely solely on the G2–M checkpoint after DNA damage, G2–M checkpoint abrogation through small-molecule inhibition of WEE1 sensitizes p53-deficient cells to DNA-damaging agents, including gemcitabine and platinum compounds (59). However, there is opposing literature as to whether WEE1 inhibition is more effective in p53 mutant backgrounds compared with p53 wild-type cancer cell lines (59–61).
Emerging data suggest that cancers with increased intrinsic replication stress (e.g., oncogene-induced, that is, arising from HPV E6/E7 and/or PARK2/FBXW7 loss-dependent cyclin E1 dysregulation) and/or attenuated DNA damage repair pathways, may exhibit higher sensitivity to cell-cycle checkpoint kinase inhibitors, such as prexaseritib (CHK1i) and berzosertib (ATRi; refs. 5, 58, 62–64). This emphasizes that there are alternative ways of reducing checkpoint function in cancer, and hence, we need to improve on our methods to call an individual tumor's checkpoint control deficiencies to improve patient stratification. Therefore, pretreatment tissues from patients should be analyzed by targeted next-generation sequencing and correlated with response.
Furthermore, predictive pharmacodynamic biomarkers, used to verify target inhibition and subsequent modulation of the downstream signaling pathways in vivo, should be monitored throughout treatment and are crucial in predicting the response (65). Biomarker levels depend on the type of chemotherapeutic agent, the concentration of the drug, and the duration of time between combination treatment and analysis of the biomarker, and could help make informed decisions about treatment continuation and optimal scheduling. A number of IHC markers, including γH2AX (a marker of DNA damage; ref. 66), p-H3 (which correlates with chromosome condensation in mitosis), and p-CDC2 (a marker of CDK inhibition), can be used to assess effectiveness across the range of combination therapies discussed here (67, 68). Recent studies have shown that increased DNA damage is responsible for a synergistic antitumor effect and correlates closely with potentiation of two drugs, as cell lines that were not sensitive did not show any additive changes in γH2AX (54). As such, γH2AX levels can help guide treatment timing as exemplified in Montano and colleagues (49). Addition of CHK1 inhibitor (MK-8776) following gemcitabine in xenografts at 42 hours led to only a 10% increase in cells staining positive for γH2AX, whereas CHK1i treatment 18 hours post-gemcitabine led to a 50% increase, suggesting that at later time-points following gemcitabine, cells have already recovered from gemcitabine-mediated replicative stress (49).
Clinical Considerations for Combination Therapy
In Table 1, we present a comprehensive survey of current clinical trials testing combinations of checkpoint inhibitors, specifically WEE1, ATM, ATR, CHK1, and CHK2, together with DNA-damaging agents (https://www.clinicaltrials.gov). Recent analysis of phase II clinical trials showed that over half of the sampled phase II studies did not cite any matched preclinical or preceding clinical evidence, suggesting that the design of clinical trials adding cell-cycle checkpoint inhibitors on top of standard of care chemo- or radiotherapy does not always rest on a sound mechanistic and biological basis (69). Here, leading on from the discussion on preclinical data, we focus on scheduling, dosing, and patient stratification in clinical trials and the lessons that can be learned from failed and successful combination therapy trials. The data we have collated demonstrate that the clinical efficacy of combining checkpoint kinase inhibitors with cytotoxic agents is influenced by the drug regimen used.
Trial name . | Phase . | Status . | Compounds . | Treatment arms . | Scheduling . | Indication . | Efficacy . | Biomarkers evaluated . | Refs . |
---|---|---|---|---|---|---|---|---|---|
NCT02157792 | I | Completed | VX-790 (M6620), Gemcitabine, Cisplatin, Etoposide, Carboplatin, Irinotecan | A. M6620 + gemcitabine (+ cisplatin) | 3 + 3 design | Part A, B, B2: Advanced solid tumors | C2/C3: | 1 | |
B. M6620 + cisplatin (+ etoposide) | Part C1: TP53-mutated NSCLC | PFS: 4.1 (90% CI, 1.6–6.9 months). | |||||||
C1. M6620 + gemcitabine | C2/C3: BRCA1/2 wild-type advanced/metastatic TNBC | OR: 38.9% (90% CI, 19.9%–60.8%) | |||||||
C2. M6620 + cisplatin | |||||||||
C3. M6620 + cisplatin/carboplatin | |||||||||
NCT02627443 | I/II | Recruiting | VX-790 (M6620), Carboplatin, Gemcitabine Hydrochloride | A. Carboplatin + Gemcitabine Hydrochloride + VX-970 | Carboplatin IV over 30 minutes on day 1, gemcitabine hydrochloride IV over 30 minutes on days 1 and 8, and ATR kinase inhibitor VX-970 IV over 60 minutes on days 2 and 9. | Metastatic ovarian, primary peritoneal, or fallopian tube cancer | |||
NCT02567409 | II | Active, not recruiting | VX-790 (M6620), Cisplatin, Gemcitabine Hydrochloride | A. Cisplatin + Gemcitabine Hydrochloride + VX-970 | Gemcitabine hydrochloride IV over 30 minutes on days 1 and 8, and cisplatin IV over 60 minutes on day 1. Patients in Arm B also receive ATR kinase inhibitor VX-790 IV over 60 minutes on days 2 and 9. | Metastatic urothelial carcinoma | TP53, CDKN1A and ERCC2 mutation analysis | ||
B. Cisplatin + Gemcitabine Hydrochloride | |||||||||
NCT02487095 | I/II | Recruiting | VX-790 (M6620), Topotecan | A. Topotecan + VX-970 | Topotecan IV on days 1 through 5. They will get VX-970 IV on day 5 alone or on day 5 and day 2. | SCLC | Phase I: | 2 | |
PR: 2/21 | |||||||||
SD: 7/21 | |||||||||
NCT02595892 | II | Active, not recruiting | VX-790 (M6620), Gemcitabine Hydrochloride | A. Gemcitabine Hydrochloride | Gemcitabine hydrochloride IV over 30 minutes on days 1 and 8 (Arm B: + ATR kinase inhibitor VX-790 IV over 60–90 minutes on days 2 and 9). | High-grade serous ovarian cancers | mPFS: 22.9 (B) vs. 14.7 (A) weeks (HR = 0.57, P = 0.044) | 3 | |
B. Gemcitabine Hydrochloride + VX-970 | |||||||||
NCT02589522 | I | Recruiting | VX-790 (M6620), WBRT | A. VX-970 + RT | A. WBRT QD 5 days a week for 15 fractions. Patients also receive VX-790 IV over 60–90 minutes twice a week, 18–30 hours after first RT. | NSCLC, SCLC, or neuroendocrine tumors | pATR T1989, pCHK1 S345 and RAD51 in (CSF) post-VX970 and ATR, CCNE amplification and DNA-PK expression status pre-treatment | ||
B. VX-970 + RT + surgery | B. VX-970 IV over 60–90 minutes 2–4 hours prior to surgery. After surgery, patients undergo WBRT and receive VX-970 as in Group A. | ||||||||
NCT02567422 | I | Recruiting | VX-790 (M6620), Cisplatin, RT | A. VX970 + Cisplatin + RT | VX-970 IV over 60 minutes on day 7 and then weekly on day 2 and cisplatin IV over 30–60 minutes weekly on day 1. Patients also undergo RT once daily, 5 days a week. | Advanced HNSCC | Induction of γH2AX post-VX970 | ||
NCT02595931 | I | Recruiting | VX-790 (M6620), Irinotecan Hydrochloride | A. Irinotecan Hydrochloride + VX-970 | Irinotecan hydrochloride IV over 90 minutes and M6620 IV over 60 minutes on days 1 and 15. | Metastatic malignant neoplasm | |||
NCT02723864 | I | Recruiting | VX-790 (M6620), Cisplatin, Veliparib | A. VX-970 + Cisplatin + Veliparib | VX-970 IV on days 2 and 9; Veliparib will be administered orally BID days 1–3 and 8–10 of each cycle; Cisplatin will be administered at 40 mg/m2 IV day 1 (and day 8 from DL3 onwards) of each cycle | Refractory solid tumors | PR: 3/22 | γH2AX, RAD51, pNbs1, and pATR in tumour biopsies at MTD (IHC) and ctDNA | 4 |
SD: 12/22 | |||||||||
NCT02264678 | I/Ib | Recruiting | Ceralasertib (AZD6738), Carboplatin, and others | A. Ceralasertib + Carboplatin. | Single dose of ceralasertib on day 1, followed by multiple dosing in combination with carboplatin. | Solid malignant tumour | Evaluation functional ATR inhibition, ctDNA and CTCs post- Ceralasertib | ||
B. Ceralasertib | (A: Advanced lung adenocarcinoma with low expression of ATM) | ||||||||
PATRIOT NCT02223923 | I | Active, not recruiting | Ceralasertib (AZD6738), Palliative RT | A. AZD6738 | A. 3 +3 design | Solid tumors | 5 | ||
B. AZD6738 + RT (head and neck) | B. Dose escalation of AZD6738 with 20 Gy in 10 fractions of RT, starting at least 2 dose levels below the currently tolerated dose of AZD6738 | ||||||||
AZD6738 + C. RT (Abdomen/Pelvis) | |||||||||
NCT02630199 | I | Recruiting | Ceralasertib (AZD6738), Paclitaxel | A. AZD6738 + Paclitaxel | AZD6738 on D1, D8, and D21 followed by combination therapy with weekly paclitaxel from cycle 1. Paclitaxel 80 mg/m2 on days 1, 8, and 15 every 4 weeks | Refractory cancer | CR: 1/53 | 6 | |
PR: 12/53 | |||||||||
SD: 18/53 | |||||||||
NCT03641313 | II | Active, not recruiting | VX-790 (M6620), Irinotecan Hydrochloride | A. Irinotecan Hydrochloride + VX-790 | Irinotecan IV over 90 minutes and ATR kinase inhibitor M6620 IV over 60 minutes on days 1 and 15. | Progressive, metastatic, or unresectable TP53-mutant gastric or gastroesophageal junction cancer | Correlative studies in the first 9 patients: γH2AX, KAP1 (p)-Ser 824 and p-ATR analysis from cycle 1 day 1 post-irinotecan biopsy and cycle 2 day 2 within 24 hours post-VX970 biopsy | ||
NCT03517969 | II | Recruiting | VX-790 (M6620), Carboplatin, Docetaxel | A. Docetaxel + Carboplatin | A. Docetaxel IV over 60 minutes and carboplatin IV over 30 minutes on day 1 or carboplatin alone on day 1. | Metastatic castration-resistant prostate cancer | |||
B. Carboplatin + VX-970 | B. Carboplatin IV over 30 minutes on day 1 and VX-790 IV over 60–90 minutes on days 2 and 9 | ||||||||
NCT03641547 | I | Recruiting | VX-790 (M6620), Cisplatin, Capecitabine, RT | A. VX-970 + RT | A. Daily 140 mg/m2 VX-790 during palliative RT | ATM- or TP53-deficient esophageal cancer & other solid cancers | Apoptosis and ATR target inhibition post-VX970 (IHC) | ||
B. VX-970 + Capecitabine + Cisplatin | B. 90 mg/m2 VX-790 24 hours post cisplatin infusion | ||||||||
NCT03669601 | I | Recruiting | Ceralasertib (AZD6738), Gemcitabine | A. Ceralasertib + Gemcitabine | IV gemcitabine on days 3, 10, and 17. Oral tablet AZD6738 once daily and intermittently for up to 12 days of a 28-day cycle | Locally advanced or metastatic tumors | |||
NCT03896503 | II | Recruiting | VX-790 (M6620), Topotecan Hydrochloride | A. Topotecan Hydrochloride | Topotecan hydrochloride IV over 30 minutes on days 1–5. (In Arm B also: VX-790 IV over 60 minutes on days 2 and 5) | SCLC and small cell cancers outside of the lungs | Expression of SLFN11, cMYC, and ATM | ||
B. Topotecan Hydrochloride + VX-790 | |||||||||
NCT03309150 | I | Active, not recruiting | VX-790 (M6620), Carboplatin, Paclitaxel | A. M6620 Monotherapy or Combination Therapy | VX-970 IV as a monotherapy on days 1, 8, and 15 OR as a combination therapy on day 2 and 9 of the study. carboplatin IV on day 1. Paclitaxel IV on day 1. | Advanced stage solid tumors | |||
NCT04052555 | Ib | Recruiting | VX-790 (M6620), RT | A. VX-790 + RT | Berzosertib intravenously (IV) over 60 minutes twice weekly. Patients also undergo RT 5 days a week for 5–6 weeks | Triple-negative and estrogen and/or progesterone receptor positive, HER2 negative breast cancer | |||
NCT04216316 | Ib/II | Not yet recruiting | Avelumab | A. Avelumab + VX-790 + Gemcitabine + Carboplatin | A. Avelumab IV over 60 minutes and gemcitabine IV over 30 minutes on days 1 and 8. Patients also receive carboplatin IV over 30 minutes on day 1 and VX-970 IV over 60 minutes on days 2 and 9. | NSCLC | ATM assay | ||
VX-790 (M6620), Carboplatin, Gemcitabine, Gemcitabine Hydrochloride | B. Avelumab + Gemcitabine + Carboplatin | B. Avelumab, Gemcitabine, and Carboplatin as in Arm A | |||||||
NCT03704467 | Ib/II | Completed | Carboplatin, Paclitaxel, Gemcitabine, M6620, Bevacizumab, Avelumab | A. Carboplatin + M6620 + Avelumab | A. 90 mg/m2 VX-790 IV on day 2, IV infusion of avelumab 1,600 mg and carboplatin on day 1 | PARPi-resistant ovarian cancer (BRCA1/2 mutation) | |||
B. SoC: Gemcitabine + Paclitaxel | B. Carboplatin IV AUC 5 (+ paclitaxel) or AUC 4 (+ gemcitabine) | ||||||||
NCT02588105 | I | Active, not recruiting | AZD0156, Olaparib, | A. AZD0156 + Olaparib | Advanced cancer | ATM assay, ctDNA, CTCs | |||
Irinotecan, Fluorouracil, Folinic Acid | B. AZD0156 + Irinotecan + Fluorouracil + Folinic Acid | ||||||||
NCT03423628 | I | Recruiting | AZD1390, RT | A. IMRT + AZD1390 | A. 35 Gy IMRT at daily fractions of 3.5 Gy over 10 fractions (2 weeks). B. 30 Gy WBRT/PBRT daily fractions of 3 Gy over 10 fractions (2 weeks). C. 60 Gy IMRT daily fractions of 2 Gy over 30 fractions (6 weeks). AZD1390: Cycle 0 (arms A and C): 1 dose prior to RT. Cycle 1 (all arms): Intermittent or continuous dosing during RT. Cycle 2 (arms A and C): 2 weeks adjuvant treatment after RT. | Recurrent glioblastoma multiforme | |||
B. WBRT/PBRT + AZD1390 | |||||||||
C. IMRT + AZD1390 | |||||||||
NCT02906059 | I | Recruiting | AZD1775, Irinotecan | A. AZD1775 + Irinotecan | Standard dose Irinotecan is given on day 1 of every 2 week cycle. AZD1775 is administered PO twice daily for 3 to 5 days of each cycle, starting cycle 2. | Second-line metastatic colorectal cancer, RAS or BRAF mutated | Adequate target engagement of Wee1, changes in markers of DNA damage, TP53 mutation status | ||
NCT02666950 | II | Completed | AZD1775, Cytarabine | A. Cytarabine + AZD1775 | 20 mg cytarabine (AraC) SC twice daily and 200 mg WEE1 inhibitor (AZD1775) PO daily on days 1–5 and days 8–12 OR receive only 200 mg WEE1 inhibitor (AZD1775) PO daily on days 1–5, 8–12, 15–19, and 22–26. | Advanced acute myeloid leukemia or myelodysplastic syndrome | |||
B. AZD1775 | |||||||||
NCT02194829 | I/II | Active, not recruiting | AZD1775, Gemcitabine Hydrochloride, Nab-paclitaxel | Phase I | Paclitaxel albumin-stabilized nanoparticle formulation IV over 30 minutes and gemcitabine hydrochloride IV over 30 minutes on days 1, 8, and 15. Patients also receive WEE1 inhibitor AZD1775 orally (PO) daily on days 1, 2, 8, 9, 15, and 16. | Metastatic pancreatic adenocarcinoma | |||
A. Paclitaxel + AZD1775 + Gemcitabine Hydrochloride (Dose Level 1) | |||||||||
B. Paclitaxel + AZD1775 + Gemcitabine Hydrochloride (Dose Level 2) | |||||||||
Phase II | |||||||||
C. Paclitaxel + Gemcitabine Hydrochloride | |||||||||
D. Paclitaxel + AZD1775 + Gemcitabine Hydrochloride | |||||||||
NCT02448329 | II | Recruiting | AZD1775, Paclitaxel | A. AZD1775 + Paclitaxel | AZD1775 225 mg BID q 12 hours (x 5 doses, 2.5 days) administered days 1–3. Weekly paclitaxel 80 mg/m2 IV on 1, 8, and 15 of a four week l cycle | TP53-mutated advanced gastric adenocarcinoma | |||
NCT03028766 | I | Recruiting | AZD1775, Cisplatin, IMRT | A. AZD1775 + Cisplatin | AZD1775 by mouth, twice a day for 3 days on days 2, 9, 23, and 30. Cisplatin 40 mg/m2 IV delivered over 1 hour on days 2, 9, 16, 23, and 30. IMRT will be delivered 5 days a week (once daily, Monday to Friday) for 6 weeks commencing within 3 months of surgery. | Head and neck cancer | 7 | ||
B. AZD1775 + Cisplatin + IMRT | |||||||||
NCT01922076 | I | Active, not recruiting | AZD1775, RT | A. AZD1775 + RT | RT 5 days a week for 6 weeks (up to 30 fractions). Patients also receive AZD1775 orally (PO) on days 1–5 of weeks 1, 3, and 5; days 1–5 of weeks 1, 3, and 5 AND days 1, 3, and 5 of weeks 2, 4, and 6; OR days 1–5 of weeks 1–6 depending on dose level assignment. | Anaplastic astrocytoma, glioblastoma, gliosarcoma, diffuse midline glioma with histone H3 K27M mutation | Change in p-CDC2, p-HH3 and γH2AX expression | ||
NCT02095132 | I/II | Active, not recruiting | AZD1775, Irinotecan | A. Irinotecan Hydrochloride + AZD1775 | Patients receive irinotecan hydrochloride PO and AZD1775 PO on days 1–5. | Relapsed or refractory solid tumors | Changes p-CDK1 level, MYC and MYCN expression, p-WEE1 levels, EZH2 and γH2AX histone levels in tumor for correlation analyses | ||
NCT03345784 | I | Recruiting | AZD1775, Cisplatin, RT | A. AZD1775 + Cisplatin + RT | Patients undergo external beam radiation therapy on days 1–5 and receive adavosertib orally (PO) on days 1, 3, and 5 or once daily (QD) on days 1–5 and cisplatin IV over 1 hour on day 1 or 3. Cycles repeat each week for up to 5 weeks. | Cervical, vaginal, or uterine cancer | (p)CDC2, Ki67, γH2AX, pH3, and CC3. | ||
NCT01849146 | I | Active, not recruiting | AZD1775, Temozolomide, RT | A. AZD1775 + Temozolomide + RT | A. Patients receive adavosertib orally (PO) on days 1, 3, and 5 or days 1–5 weekly and temozolomide PO once daily (QD) for 6 weeks. Patients also undergo concurrent RT 5 days per week for 6 weeks. | Newly diagnosed or recurrent glioblastoma | pRb (S807/811), Ki67, p-CDC2, P-gp, cleaved caspase (apoposis), pWee1 expression levels, TP53 mutation status, MGMT methylation | 8 | |
B. AZD1775 + Temozolomide | B. Patients receive adavosertib PO QD on days 1, 3, and 5 or 1–5 weekly, and temozolomide PO QD on days 1–5. Treatment repeats every 28 days for up to 6 cycles. | ||||||||
NCT02272790 | II | Active, not recruiting | AZD1775, Gemcitabine, Paclitaxel, and others | A. AZD1775 + Gemcitabine | A. AZD1775 (175 mg PO) will be taken on Days 1–2, 8–9, and 15–16. Gemcitabine 800 mg/m² will be administered IV on days 1, 8, and 15 of each 28 day cycle. | TP53-mutated | mPFS (Arm C): 10.1 months | 9 | |
B. AZD1775 + Paclitaxel | B. Five doses of AZD1775 (225 mg PO BID) will be taken in approximate 12 hour intervals over 2.5 days weekly (Days 1–3, 8–10, and 15–17). Weekly paclitaxel 80 mg/m² IV will be administered on Day 1, 8, and 15 of each 28 day cycle. | Platinum-Resistant Epithelial Ovarian, Fallopian Tube, or Primary Peritoneal Cancer | ORR: 1/9 (A), 11/38 (B), 7/23 (C), 8/12 (C2) | ||||||
C/C2. AZD1775 + Carboplatin | C. Adavosertib 225 mg orally BID (5 doses over 3 days) on Days 1–3 of 21 day cycles. Carboplatin AUC 5 IV on Day 1 of 21 day cycles. | mPFS: 1.7. (A), 5.5 (B), 4.2 (C), and 12.0 (C2) months | |||||||
C2. Adavosertib 225 mg orally BID (5 doses over 3 days) on Days 1–3, 8–10, and 15–17 of 21 day cycles. Carboplatin AUC 5 IV on Day 1 of 21 day cycles. | |||||||||
NCT02937818 | II | Active, not recruiting | AZD1775, Carboplatin and others | A. AZD1775 + Carboplatin | AZD1775 twice daily (oral) for 2.5 days from Day 1 + CBDP area under the curve 5 (Day 1) | Platinum Refractory Extensive-Stage Small-Cell Lung Cancer | |||
NCT01164995 | II | Unknown | AZD1775, Carboplatin | A. AZD1775 + Carboplatin | Carboplatin AUC5 (IV. 30 min) at day 1. Concomitantly with the start of the carboplatin infusion 225 mg of AZD1775 will be administered as an oral capsule, followed by 4 additional doses at 12 hour increments (= 5 BID doses of AZD1775 in 2.5 days in total). | TP53-mutated Epithelial Ovarian Cancer | ORR: 43% | p-CDC2 levels on Day 1 and Day 3 post-AZD1775 | 10 |
mPFS: 5.3 months | |||||||||
OS: 12.6 months | |||||||||
NCT02341456 | Ib | Completed | AZD1775, Paclitaxel, Carboplatin | A. AZD1775 + Paclitaxel + Carboplatin | AZD1775 will be administered orally as a single dose on Day 1 Cycle 0. Following a 5±2 days washout period, AZD1775 (5 doses BID over 2.5 days) will be taken in combination with paclitaxel and carboplatin in each 21-day cycle for 6 cycles. Following 6 cycles of combination treatment, patients may continue on AZD1775 monotherapy (5 doses BID Day 1 to Day 2.5 in each 21-day cycle) at the investigator's discretion. | Advanced Solid Tumours | PR: 16.7% (2/12) | 11 | |
NCT02037230 | I/II | Completed | AZD1775, Gemcitabine, Radiotherapy | A. AZD1775 + Gemcitabine + Radiation Therapy | AZD1775 will be given as an oral capsule on days 1 and 2, and on days 8 and 9. Gemcitabine 1000 mg/m2 will be infused over 30 minutes on days 1 and 8. 52.5 Gy in 25 fractions (2.1 Gy/fraction), using intensity modulated radiation therapy (IMRT). Radiation therapy will be administered after chemotherapy. | Unresectable Adenocarcinoma of the Pancreas | mPFS: 9.4 months | p-CDC2 (IHC) | 12 |
OS: 21.7 months | |||||||||
NCT01357161 | II | Completed | AZD1775, Paclitaxel, Carboplatin | A. AZD1775 225 mg + Paclitaxel + Carboplatin | A. 225 mg AZD1775 twice daily (BID) starting on Day 1 of Cycle 1 (cycle = 21 days) for a total of 5 doses. Participants receive AZD1775 in combination with paclitaxel (175 mg/m2) and carboplatin (area under the curve [AUC] 5). (Arm B: Placebo instead of AZD1775) | TP53-mutated non-low grade, non-borderline ovarian, fallopian tube, or primary peritoneal cancer which has progressed after paclitaxel/platinum-based therapy | mPFS: 7.9 (A) vs. 7.3 months (B) | TP53 mutation subtypes | 13, 30 |
B. Placebo + Paclitaxel + Carboplatin | (HR = 0.63, P = 0.080 | ||||||||
ORR: 1.4% vs. 75.8% (P = 0.459) | |||||||||
NCT02513563 | II | Recruiting | AZD1775, Carboplatin, Paclitaxel | A. AZD1775 + Carboplatin + Paclitaxel | IV carboplatin AUC 5, IV paclitaxel 175 mg/m2, and oral AZD1775 225 mg twice/day for 2.5 days every 21 days for 4–6 cycles followed by maintenance AZD1775 at the same doses | Squamous Cell Lung Cancer | PR: 30% (3/10) | Correlative analyses of p53, PAXIP1, and WEE1 | 14 |
SD: 50% (5/10) | |||||||||
NCT02508246 | I | Completed | AZD1775, Cisplatin, Docetaxel | A. AZD1775 + Cisplatin + Docetaxel | WEE1 inhibitor AZD1775 orally (PO) twice daily (BID) on days 2–4, 9–11, and 16–18, and day -7 prior to course 1, day 1 for PD assessment. Patients also receive cisplatin IV on days 1 (or up to two days after last dose of WEE1 inhibitor AZD1775 lead-in is completed), 8 (or 7 days after first chemotherapy dose), and 15, and docetaxel IV on days 1, 8, and 15. | Squamous Cell Carcinoma of the Head and Neck (HNSCC) | CR: 2/10 | TP53 mutation status, HPV status, WEE1 and p-CDC2 levels | 15 |
PR: 5/10 | |||||||||
NCT02101775 | II | Active, not recruiting | AZD1775, Gemcitabine Hydrochloride | A. AZD1775 + Gemcitabine Hydrochloride | A. Patients receive WEE1 inhibitor AZD1775 orally (PO) on days 1, 2, 8, 9, 15, and 16 and gemcitabine hydrochloride intravenously (IV) over 30 minutes on days 1, 8, and 15. Courses repeat every 28 day. (Arm B: Placebo instead of AZD1775) | Recurrent Ovarian, Primary Peritoneal, or Fallopian Tube Cancer | pCDC2 and γH2AX levels, TP53 mutations | ||
B. Placebo + Gemcitabine Hydrochloride | |||||||||
NCT00648648 | I | Completed | AZD1775, Cisplatin, Carboplatin, Gemcitabine | A. AZD1775 + Gemcitabine | 1) AZD1775 +Gemcitabine (1,000 mg/m2), 2) AZD1775 + Cisplatin (75 mg/m2), or 3) AZD1775 +Carboplatin at an area under the time curve concentration of 5 mg/min/mL (AUC5). Following completion of Part 2-A, AZD1775 will be administered twice daily (BID) for 2.5 days (multi-dose) starting concomitantly with each administration of chemotherapy in Part 2-B. | Advanced solid tumors | SD: 53% (94/176) | pCDC2 24 and 48 hours post-AZD1775 | 16 |
B. AZD1775 + Cisplatin | PR: 10% (17/176) | ||||||||
C. AZD1775 + Carboplatin | |||||||||
NCT03012477 | II | Active, not recruiting | Cisplatin | A. Cisplatin + AZD1775 | Cisplatin will be given on the first day of every cycle. AZD1775 will be administered in clinic on the first day of the second cycle. Subsequent doses will be taken approximately 12 hours apart for a total of five doses. | Breast cancer | |||
NCT02585973 | Ib | Active, not recruiting | AZD1775, Cisplatin, IMRT | A. AZD1775 + Cisplatin + IMRT | AZD1775 given twice daily (BID) for three consecutive days (M-W) concomitantly with standard of care cisplatin (40 mg/m2 IV infused over 1-hour D1 of each week of radiation) and radiation (Total dose will be 70 Gy at 2 Gy/fx, 35 fractions, Mon to Fri, for 7 weeks) | HNSCC of the oropharynx, larynx, hypopharynx, or oral cavity | p-CDC2 and γH2AX levels, TP53 mutation analysis | ||
NCT02797977 | I/II | Active, not recruiting | SRA737, Gemcitabine, Cisplatin | A. SRA737 + Gemcitabine + Cisplatin | SRA737 will be administered orally on Days 2, 3, 9, 10, 16, and 17 of each 28-day cycle. Subjects will receive a single dose of SRA737 between 4 to 7 days prior to starting the first cycle. Gemcitabine will be administered intravenously on Days 1, 8, and 15 of each 28-day cycle. | HGSOC, SCLC, STS, and cervical/anogenital cancer | |||
NCT04023669 | I | Recruiting | Prexasertib (LY2606368), Gemcitabine, Cyclophosphamide | A. Prexasertib + Cyclophosphamide | A. Cyclophosphamide IV on days 1 and 15 and prexasertib IV on days 2 and 16. Cycles repeat every 28 days for up to 24 months (26 cycles). | Medulloblastoma | |||
B. Prexasertib + Gemcitabine | B. Gemcitabine IV on days 1 and 15 and prexasertib IV on days 2 and 16. Cycles repeat every 28 days for up to 24 months (26 cycles). | ||||||||
NCT01139775 | I/II | Completed | Pemetrexed, Prexasertib (LY2606368), Cisplatin | A. Pemetrexed + Prexasertib + Cisplatin | Pemetrexed 500 milligrams per meter square (mg/m2) + cisplatin 75 mg/m2 on Day 1 (Arm A: LY2603618 at 130–275 mg on Day 2) | Nonsquamous NSCLC | mPFS: 4.7 (A) vs. 1.5 months (B (P = 0.022) | 17, 18 | |
B. Pemetrexed + Cisplatin | |||||||||
NCT01870596 | II | Completed | SCH 900776, Cytarabine | A. SCH 900776 + Cytarabine | Patients receive cytarabine IV continuously over 72 hours on days 1–3 and 10–12 and Chk1 inhibitor SCH 900776 IV over 30 minutes on days 2, 3, 11, and 12. | Acute Myeloid Leukemia | CR: 36% (5/14) (A) vs. 44% (8/18) (B) | 19 | |
B. Cytarabine | PR: 7% (1/14) vs. 6% (1/14) (B) | ||||||||
mPFS: 5.9 vs. 4.1 months, p = ns) | |||||||||
NCT02649764 | I | Active, not recruiting | Cytarabine, Prexasertib (LY2606368), Fludarabine Phosphate | A. Cytarabine + Prexasertib + Fludarabine Phosphate | Patients ≤65 years of age: receive fludarabine IV over approximately 2 hours on days 1–4, cytarabine IV over 4 hours on days 1–4, and prexasertib (LY2606368) IV over approximately 2 hours on days 1, 3, and 4 or on days 1–4. Patients > 65 years of age: receive fludarabine IV over approximately 2 hours on days 1–3, cytarabine IV over 4 hours on days 1–3, and prexasertib (LY2606368) IV over approximately 2 hours on days 1, 3, and 4 or on days 1–4. Treatment for both age groups repeats every 28 days for up to 5 courses. | Relapsed or refractory (first or second salvage) AML or high-risk myelodysplastic syndrome (HRMDS). | Phosphorylated H2AX, Chk1/2, Cdc25, Rb, Akt, and CDC2 levels (WB or Flow Cytometry) | ||
NCT02124148 | I | Completed | Prexasertib (LY2606368), Cisplatin, Pemetrexed, Fluorouracil, and others | A/A2/A3. Prexasertib + Cisplatin | A. Prexasertib and cisplatin administered IV once every 21 days. | Advanced Cancer | CR: 0% (A), 0% (A2), 5% (1/19) (A3) | 20 | |
C. Prexasertib + Pemetrexed | A2: Prexasertib and cisplatin administered IV every 21 days; G-CSF administered subcutaneously (SC) starting approximately 24 hours after each prexasertib dose every 21 days. | Part B: KRAS wild-type colorectal cancer | PR: 21% (3/9) (A), 0% (A2), 11% (2/19) (A3) | ||||||
D. Leucovorin + 5-FU + Prexasertib | Part A3: Cisplatin administered IV on day one and prexasertib administered IV on day two once every 21 days. | SD: 14% (2/9) (A), 50% (12/24) (A2), 37% (7/19) (A3) | |||||||
Part C: Pemetrexed administered IV on day one and prexasertib administered IV on day one and two every 21 days. | |||||||||
Part D: Leucovorin administered IV on day one, 5-FU administered IV bolus on day one and by continuous IV on days one to three (46 hours), and prexasertib administered IV on day three every 14 days | |||||||||
NCT01341457 | I | Completed | LY2603618, Gemcitabine | A. 170 mg LY2603618 + Gemcitabine | Gemcitabine 1,000 mg/m2 IV on days 1, 8, and 15. 170 or 230 mg LY2603618 administered intravenously on days 2, 9, and 16 of at least one 28-day cycle. | Solid advanced or metastatic tumors | Best OR: 2/7 (A), 5/10 (B) | ||
B. 230 mg LY2603618 + Gemcitabine | |||||||||
NCT00839332 | I/II | Completed | LY2603618, Gemcitabine | A. LY2603618, Gemcitabine | 1,000 mg/m2 gemcitabine as a 30-minute continuous IV infusion once per week 24 hours prior to LY2603618. | Pancreatic cancer | OS: 7.8 (A) vs. 8.3 months (B) | 21 | |
B. Gemcitabine | Phase II: 230 mg LY2603618 as a 1-hour continuous IV infusion once per week for 3 weeks, followed by 1 week of rest. | ||||||||
NCT00988858 | II | Completed | LY2603618, Pemetrexed | 500 mg/m2 Pemetrexed IV on Day 1 and 150 mg/m2 IV on day 2 | Advanced or metastatic NSCLC | PR: 9.1% (5/55) | 22 | ||
SD: 36.% (20/50) | |||||||||
mPFS: 2.3 months | |||||||||
NCT04095221 | I/II | Recruiting | Prexasertib (LY2606368), Irinotecan | A. Prexasertib + Irinotecan | 3 + 3 design | Relapsed or Refractory Desmoplastic Small Round Cell Tumor and Rhabdomyosarcoma | |||
NCT02555644 | I | Completed | Prexasertib (LY2606368), Cisplatin, Cetuximab, IMRT | A. Prexasertib + Cisplatin + Radiation Therapy | A. Prexasertib administered IV every 14 days. Cisplatin administered IV every 7 days. IMRT administered 5 days per week. | HNSCC of the oropharynx, hypopharynx, or larynx, or SCC of the anus | |||
B. Prexasertib + Cetuximab + Radiation Therapy | B. Prexasertib administered IV every 14 days. Cetuximab administered IV every 7 days. IMRT administered 5 days per week. | ||||||||
NCT00779584 | I | Completed | MK-8776, (SCH 900776) | A. AZD1775 | IV infusion with MK-8776 at seven dose levels ranging from 10 to 150 mg/m2 as monotherapy and then in combination with gemcitabine 800 mg/m2 (part A, n = 26) or gemcitabine 1,000 mg/m2 (part B, n = 17). | Advanced solid tumor malignancy or lymphoma | PR: 7% (2/30) | 23 | |
Gemcitabine | B. AZD1775 + Gemcitabine | SD: 43% (13/30) (A) | |||||||
NCT00045513 | I/II | Completed | UCN-01, Fludarabine Phosphate | A. UCN-01 + Fludarabine Phosphate | Patients receive UCN-01 IV over 3 hours on day 1 and fludarabine IV over 30–60 minutes on days 1–5. | Chronic Lymphocytic Leukemia or Lymphocytic Lymphoma | |||
NCT00006464 | I | Completed | UCN-01, Cisplatin | A. Cisplatin + UCN-01 | Patients receive cisplatin IV over 1 hour on day 1 and UCN-01 IV continuously over 36–72 hours beginning on day 2. | Advanced or metastatic solid tumor incurable by surgery or other standard therapy | |||
NCT00700336 | I/II | Completed | Pemetrexed, Cisplatin and CBP501 | A. Pemetrexed + Cisplatin + CBP501 | Advanced solid tumors and in chemotherapy-naïve patients with malignant pleural mesothelioma | ||||
B. Pemetrexed + Cisplatin | |||||||||
NCT00551512 | I | Completed | Cisplatin, CBP501 | A. CBP501 | CBP501 alone (D1/D8/D15, from 0.9 mg/m2), or with Cisplatin (both on D1, from 3.6 mg/m2 CBP501, 50 mg/m2 cisplatin). | Advanced refractory solid tumors (Arm B expansion: synchronous ovarian and endometrial carcinoma) | PR: 0/30 (A), 2/14 (B) | Rad51, BRCA1, ATM, FANCD2 and MUS81, XPF and Polη, ATM and phospho-MAPKAP-K2 levels (IHC) | 24 |
B. Cisplatin + CBP501 | SD: 7/30 (A), 5/14 (B) | ||||||||
NCT00413686 | I | Completed | AZD7762, Gemcitabine | In the first cycle (cycle 0), patients received a single dose of AZD7762 administered as a 60-minutes IV infusion on days 1 and 8 of a 14-day run-in cycle. In subsequent cycles, patients received AZD7762 at the same dose as cycle 0 in combination with 750 or 1,000 mg/m2 gemcitabine administered as a 30-minute IV infusion. The first seven patients received combination treatment on days 1, 8, and 15 of a 28-day cycle. | Advanced Solid Malignancies | PR: 1/38 (AZD7762 6 mg/gemcitabine 750 mg/m2 and AZD7762 9 mg cohort) | 25 | ||
SD: 5/38 (AZD7762 6 mg/gemcitabine 750 mg/m2, AZD7762 14 mg and AZD7762 30 mg cohorts, n = 1 patient per cohort, and AZD7762 40 mg cohort n = 2 patients) | |||||||||
NCT00031681 | I | Completed | UCN-01, Irinotecan Hydrochloride | A. UCN-01 + Irinotecan Hydrochloride | Patients receive irinotecan hydrochloride intravenously (IV) over 90 minutes on days 1, 8, 15, and 22 and 7-hydroxystaurosporine IV over 3 hours on days 2 and 23. | Metastatic or Unresectable Solid Tumors or Triple Negative Breast cancer | PR: 2/25 | TP53 mutation, p-S6, p-CDC2, p-Chk1(S317), p-HH3, γH2AX and cleaved caspase 3 (IHC) | 26 |
SD: 12/25 | |||||||||
NCT00036777 | I | Completed | UCN-01, Carboplatin | A. UCN-01 + Carboplatin | Patients receive carboplatin IV over 1 hour followed by UCN-01 IV over 3 hours on day 1. | Advanced solid tumors | |||
NCT00004263 | I | Completed | UCN-01, Cytarabine | A. UCN-01 + Cytarabine | Patients receive cytarabine IV over 24 hours on days 1–4 of each course. Patients receive UCN-01 IV over 24 hours on days 2–4 of course 1 and over 36 hours beginning on day 2 of subsequent courses. | Refractory or relapsed acute myelogenous leukemia or myelodysplastic syndrome | SD: 2/11 | Akt, p-Akt(S473), and p38α | 27 |
MAPK levels (IHC) | |||||||||
NCT00039403 | I | Completed | UCN-01, Gemcitabine Hydrochloride | A. UCN-01 + Gemcitabine Hydrochloride | Patients receive gemcitabine IV over 1–2 hours on days 1 and 8 followed by UCN-01 IV over 3 hours on day 1. | Adenocarcinoma of the pancreas | |||
NCT00098956 | II | Completed | UCN-01, Topotecan | A. UCN-01 + Topotecan | Patients receive topotecan IV over 30 minutes on days 1–5 and UCN-01 IV over 3 hours on day 1. | Relapsed or progressed SCLC | |||
NCT00045747 | II | Completed | UCN-01, Fluorouracil | A. UCN-01 + Fluorouracil | Patients receive fluorouracil IV over 24 hours on days 1, 8, 15, and 22. Patients also receive UCN-01 IV continuously over 72 hours (course 1 only) beginning on day 2. In subsequent courses, UCN-01 is infused over 36 hours. | Gemcitabine-refractory metastatic pancreatic cancer | |||
NCT00045500 | I | Completed | UCN-01, Prednisone | A. UCN-01 + Prednisone | Patients receive oral prednisone daily on days 1–5 and UCN-01 IV over 36–72 hours on days 3–5. | Refractory solid tumors or lymphomas | |||
NCT00047242 | I | Completed | UCN-01, Irinotecan Hydrochloride | A. UCN-01 + Irinotecan Hydrochloride | Patients receive UCN-01 IV over 3 hours on day 1 and irinotecan IV over 90 minutes on days 1 and 8 | Solid tumors | SD: 4/16 | 28 | |
NCT00042861 | I | Completed | UCN-01, Fluorouracil | A. UCN-01 + Fluorouracil | Patients receive leucovorin calcium (CF) IV over 2 hours and 5-FU IV (at the midpoint of CF administration) on day 1, followed by UCN-01 IV over 36–72 hours, on weeks 1–3. | Metastatic or unresectable solid tumors | |||
NCT00072267 | II | Completed | UCN-01, Topotecan Hydrochloride | A. UCN-01 + Topotecan Hydrochloride | Patients receive UCN-01 IV over 3 hours on day 1 and topotecan IV over 30 minutes on days 1–5. | Recurrent, persistent, or progressive advanced ovarian epithelial, primary peritoneal, or fallopian tube cancer | |||
NCT00019838 | I | Completed | UCN-01, Fludarabine Phosphate | A. UCN-01 + Fludarabine Phosphate | Patients receive UCN-01 IV over 72 hours on days 1–3 alone during course 1 and over 36 hours on days 1–2 during courses 2–7. Patients also receive fludarabine IV over 30 minutes beginning on day 1 and continuing for up to 5 days during courses 2–7. | Leukemia | OR: 38% | Apoptosis | 29 |
Lymphoma | CR: 1/18 | ||||||||
PR: 6/18 | |||||||||
SD: 7/18 | |||||||||
NCT00004059 | I | Completed | UCN-01, Fluorouracil | A. UCN-01 + Fluorouracil | Patients receive fluorouracil IV over 24 hours on days 1, 8, 15, and 22. Patients receive an initial dose of UCN-01 IV over 72 hours beginning on day 2 during course 1 and then maintenance UCN-01 IV over 36 hours beginning on day 2 during subsequent courses. | Advanced or refractory solid tumors | |||
NCT00045175 | I | Completed | UCN-01, Topotecan Hydrochloride | A. UCN-01 + Topotecan Hydrochloride | Patients receive UCN-01 IV over 3 hours on day 1 and topotecan IV over 30 minutes on days 1–5. | Fallopian tube cancer | |||
Ovarian cancer | |||||||||
Primary peritoneal cavity cancer |
Trial name . | Phase . | Status . | Compounds . | Treatment arms . | Scheduling . | Indication . | Efficacy . | Biomarkers evaluated . | Refs . |
---|---|---|---|---|---|---|---|---|---|
NCT02157792 | I | Completed | VX-790 (M6620), Gemcitabine, Cisplatin, Etoposide, Carboplatin, Irinotecan | A. M6620 + gemcitabine (+ cisplatin) | 3 + 3 design | Part A, B, B2: Advanced solid tumors | C2/C3: | 1 | |
B. M6620 + cisplatin (+ etoposide) | Part C1: TP53-mutated NSCLC | PFS: 4.1 (90% CI, 1.6–6.9 months). | |||||||
C1. M6620 + gemcitabine | C2/C3: BRCA1/2 wild-type advanced/metastatic TNBC | OR: 38.9% (90% CI, 19.9%–60.8%) | |||||||
C2. M6620 + cisplatin | |||||||||
C3. M6620 + cisplatin/carboplatin | |||||||||
NCT02627443 | I/II | Recruiting | VX-790 (M6620), Carboplatin, Gemcitabine Hydrochloride | A. Carboplatin + Gemcitabine Hydrochloride + VX-970 | Carboplatin IV over 30 minutes on day 1, gemcitabine hydrochloride IV over 30 minutes on days 1 and 8, and ATR kinase inhibitor VX-970 IV over 60 minutes on days 2 and 9. | Metastatic ovarian, primary peritoneal, or fallopian tube cancer | |||
NCT02567409 | II | Active, not recruiting | VX-790 (M6620), Cisplatin, Gemcitabine Hydrochloride | A. Cisplatin + Gemcitabine Hydrochloride + VX-970 | Gemcitabine hydrochloride IV over 30 minutes on days 1 and 8, and cisplatin IV over 60 minutes on day 1. Patients in Arm B also receive ATR kinase inhibitor VX-790 IV over 60 minutes on days 2 and 9. | Metastatic urothelial carcinoma | TP53, CDKN1A and ERCC2 mutation analysis | ||
B. Cisplatin + Gemcitabine Hydrochloride | |||||||||
NCT02487095 | I/II | Recruiting | VX-790 (M6620), Topotecan | A. Topotecan + VX-970 | Topotecan IV on days 1 through 5. They will get VX-970 IV on day 5 alone or on day 5 and day 2. | SCLC | Phase I: | 2 | |
PR: 2/21 | |||||||||
SD: 7/21 | |||||||||
NCT02595892 | II | Active, not recruiting | VX-790 (M6620), Gemcitabine Hydrochloride | A. Gemcitabine Hydrochloride | Gemcitabine hydrochloride IV over 30 minutes on days 1 and 8 (Arm B: + ATR kinase inhibitor VX-790 IV over 60–90 minutes on days 2 and 9). | High-grade serous ovarian cancers | mPFS: 22.9 (B) vs. 14.7 (A) weeks (HR = 0.57, P = 0.044) | 3 | |
B. Gemcitabine Hydrochloride + VX-970 | |||||||||
NCT02589522 | I | Recruiting | VX-790 (M6620), WBRT | A. VX-970 + RT | A. WBRT QD 5 days a week for 15 fractions. Patients also receive VX-790 IV over 60–90 minutes twice a week, 18–30 hours after first RT. | NSCLC, SCLC, or neuroendocrine tumors | pATR T1989, pCHK1 S345 and RAD51 in (CSF) post-VX970 and ATR, CCNE amplification and DNA-PK expression status pre-treatment | ||
B. VX-970 + RT + surgery | B. VX-970 IV over 60–90 minutes 2–4 hours prior to surgery. After surgery, patients undergo WBRT and receive VX-970 as in Group A. | ||||||||
NCT02567422 | I | Recruiting | VX-790 (M6620), Cisplatin, RT | A. VX970 + Cisplatin + RT | VX-970 IV over 60 minutes on day 7 and then weekly on day 2 and cisplatin IV over 30–60 minutes weekly on day 1. Patients also undergo RT once daily, 5 days a week. | Advanced HNSCC | Induction of γH2AX post-VX970 | ||
NCT02595931 | I | Recruiting | VX-790 (M6620), Irinotecan Hydrochloride | A. Irinotecan Hydrochloride + VX-970 | Irinotecan hydrochloride IV over 90 minutes and M6620 IV over 60 minutes on days 1 and 15. | Metastatic malignant neoplasm | |||
NCT02723864 | I | Recruiting | VX-790 (M6620), Cisplatin, Veliparib | A. VX-970 + Cisplatin + Veliparib | VX-970 IV on days 2 and 9; Veliparib will be administered orally BID days 1–3 and 8–10 of each cycle; Cisplatin will be administered at 40 mg/m2 IV day 1 (and day 8 from DL3 onwards) of each cycle | Refractory solid tumors | PR: 3/22 | γH2AX, RAD51, pNbs1, and pATR in tumour biopsies at MTD (IHC) and ctDNA | 4 |
SD: 12/22 | |||||||||
NCT02264678 | I/Ib | Recruiting | Ceralasertib (AZD6738), Carboplatin, and others | A. Ceralasertib + Carboplatin. | Single dose of ceralasertib on day 1, followed by multiple dosing in combination with carboplatin. | Solid malignant tumour | Evaluation functional ATR inhibition, ctDNA and CTCs post- Ceralasertib | ||
B. Ceralasertib | (A: Advanced lung adenocarcinoma with low expression of ATM) | ||||||||
PATRIOT NCT02223923 | I | Active, not recruiting | Ceralasertib (AZD6738), Palliative RT | A. AZD6738 | A. 3 +3 design | Solid tumors | 5 | ||
B. AZD6738 + RT (head and neck) | B. Dose escalation of AZD6738 with 20 Gy in 10 fractions of RT, starting at least 2 dose levels below the currently tolerated dose of AZD6738 | ||||||||
AZD6738 + C. RT (Abdomen/Pelvis) | |||||||||
NCT02630199 | I | Recruiting | Ceralasertib (AZD6738), Paclitaxel | A. AZD6738 + Paclitaxel | AZD6738 on D1, D8, and D21 followed by combination therapy with weekly paclitaxel from cycle 1. Paclitaxel 80 mg/m2 on days 1, 8, and 15 every 4 weeks | Refractory cancer | CR: 1/53 | 6 | |
PR: 12/53 | |||||||||
SD: 18/53 | |||||||||
NCT03641313 | II | Active, not recruiting | VX-790 (M6620), Irinotecan Hydrochloride | A. Irinotecan Hydrochloride + VX-790 | Irinotecan IV over 90 minutes and ATR kinase inhibitor M6620 IV over 60 minutes on days 1 and 15. | Progressive, metastatic, or unresectable TP53-mutant gastric or gastroesophageal junction cancer | Correlative studies in the first 9 patients: γH2AX, KAP1 (p)-Ser 824 and p-ATR analysis from cycle 1 day 1 post-irinotecan biopsy and cycle 2 day 2 within 24 hours post-VX970 biopsy | ||
NCT03517969 | II | Recruiting | VX-790 (M6620), Carboplatin, Docetaxel | A. Docetaxel + Carboplatin | A. Docetaxel IV over 60 minutes and carboplatin IV over 30 minutes on day 1 or carboplatin alone on day 1. | Metastatic castration-resistant prostate cancer | |||
B. Carboplatin + VX-970 | B. Carboplatin IV over 30 minutes on day 1 and VX-790 IV over 60–90 minutes on days 2 and 9 | ||||||||
NCT03641547 | I | Recruiting | VX-790 (M6620), Cisplatin, Capecitabine, RT | A. VX-970 + RT | A. Daily 140 mg/m2 VX-790 during palliative RT | ATM- or TP53-deficient esophageal cancer & other solid cancers | Apoptosis and ATR target inhibition post-VX970 (IHC) | ||
B. VX-970 + Capecitabine + Cisplatin | B. 90 mg/m2 VX-790 24 hours post cisplatin infusion | ||||||||
NCT03669601 | I | Recruiting | Ceralasertib (AZD6738), Gemcitabine | A. Ceralasertib + Gemcitabine | IV gemcitabine on days 3, 10, and 17. Oral tablet AZD6738 once daily and intermittently for up to 12 days of a 28-day cycle | Locally advanced or metastatic tumors | |||
NCT03896503 | II | Recruiting | VX-790 (M6620), Topotecan Hydrochloride | A. Topotecan Hydrochloride | Topotecan hydrochloride IV over 30 minutes on days 1–5. (In Arm B also: VX-790 IV over 60 minutes on days 2 and 5) | SCLC and small cell cancers outside of the lungs | Expression of SLFN11, cMYC, and ATM | ||
B. Topotecan Hydrochloride + VX-790 | |||||||||
NCT03309150 | I | Active, not recruiting | VX-790 (M6620), Carboplatin, Paclitaxel | A. M6620 Monotherapy or Combination Therapy | VX-970 IV as a monotherapy on days 1, 8, and 15 OR as a combination therapy on day 2 and 9 of the study. carboplatin IV on day 1. Paclitaxel IV on day 1. | Advanced stage solid tumors | |||
NCT04052555 | Ib | Recruiting | VX-790 (M6620), RT | A. VX-790 + RT | Berzosertib intravenously (IV) over 60 minutes twice weekly. Patients also undergo RT 5 days a week for 5–6 weeks | Triple-negative and estrogen and/or progesterone receptor positive, HER2 negative breast cancer | |||
NCT04216316 | Ib/II | Not yet recruiting | Avelumab | A. Avelumab + VX-790 + Gemcitabine + Carboplatin | A. Avelumab IV over 60 minutes and gemcitabine IV over 30 minutes on days 1 and 8. Patients also receive carboplatin IV over 30 minutes on day 1 and VX-970 IV over 60 minutes on days 2 and 9. | NSCLC | ATM assay | ||
VX-790 (M6620), Carboplatin, Gemcitabine, Gemcitabine Hydrochloride | B. Avelumab + Gemcitabine + Carboplatin | B. Avelumab, Gemcitabine, and Carboplatin as in Arm A | |||||||
NCT03704467 | Ib/II | Completed | Carboplatin, Paclitaxel, Gemcitabine, M6620, Bevacizumab, Avelumab | A. Carboplatin + M6620 + Avelumab | A. 90 mg/m2 VX-790 IV on day 2, IV infusion of avelumab 1,600 mg and carboplatin on day 1 | PARPi-resistant ovarian cancer (BRCA1/2 mutation) | |||
B. SoC: Gemcitabine + Paclitaxel | B. Carboplatin IV AUC 5 (+ paclitaxel) or AUC 4 (+ gemcitabine) | ||||||||
NCT02588105 | I | Active, not recruiting | AZD0156, Olaparib, | A. AZD0156 + Olaparib | Advanced cancer | ATM assay, ctDNA, CTCs | |||
Irinotecan, Fluorouracil, Folinic Acid | B. AZD0156 + Irinotecan + Fluorouracil + Folinic Acid | ||||||||
NCT03423628 | I | Recruiting | AZD1390, RT | A. IMRT + AZD1390 | A. 35 Gy IMRT at daily fractions of 3.5 Gy over 10 fractions (2 weeks). B. 30 Gy WBRT/PBRT daily fractions of 3 Gy over 10 fractions (2 weeks). C. 60 Gy IMRT daily fractions of 2 Gy over 30 fractions (6 weeks). AZD1390: Cycle 0 (arms A and C): 1 dose prior to RT. Cycle 1 (all arms): Intermittent or continuous dosing during RT. Cycle 2 (arms A and C): 2 weeks adjuvant treatment after RT. | Recurrent glioblastoma multiforme | |||
B. WBRT/PBRT + AZD1390 | |||||||||
C. IMRT + AZD1390 | |||||||||
NCT02906059 | I | Recruiting | AZD1775, Irinotecan | A. AZD1775 + Irinotecan | Standard dose Irinotecan is given on day 1 of every 2 week cycle. AZD1775 is administered PO twice daily for 3 to 5 days of each cycle, starting cycle 2. | Second-line metastatic colorectal cancer, RAS or BRAF mutated | Adequate target engagement of Wee1, changes in markers of DNA damage, TP53 mutation status | ||
NCT02666950 | II | Completed | AZD1775, Cytarabine | A. Cytarabine + AZD1775 | 20 mg cytarabine (AraC) SC twice daily and 200 mg WEE1 inhibitor (AZD1775) PO daily on days 1–5 and days 8–12 OR receive only 200 mg WEE1 inhibitor (AZD1775) PO daily on days 1–5, 8–12, 15–19, and 22–26. | Advanced acute myeloid leukemia or myelodysplastic syndrome | |||
B. AZD1775 | |||||||||
NCT02194829 | I/II | Active, not recruiting | AZD1775, Gemcitabine Hydrochloride, Nab-paclitaxel | Phase I | Paclitaxel albumin-stabilized nanoparticle formulation IV over 30 minutes and gemcitabine hydrochloride IV over 30 minutes on days 1, 8, and 15. Patients also receive WEE1 inhibitor AZD1775 orally (PO) daily on days 1, 2, 8, 9, 15, and 16. | Metastatic pancreatic adenocarcinoma | |||
A. Paclitaxel + AZD1775 + Gemcitabine Hydrochloride (Dose Level 1) | |||||||||
B. Paclitaxel + AZD1775 + Gemcitabine Hydrochloride (Dose Level 2) | |||||||||
Phase II | |||||||||
C. Paclitaxel + Gemcitabine Hydrochloride | |||||||||
D. Paclitaxel + AZD1775 + Gemcitabine Hydrochloride | |||||||||
NCT02448329 | II | Recruiting | AZD1775, Paclitaxel | A. AZD1775 + Paclitaxel | AZD1775 225 mg BID q 12 hours (x 5 doses, 2.5 days) administered days 1–3. Weekly paclitaxel 80 mg/m2 IV on 1, 8, and 15 of a four week l cycle | TP53-mutated advanced gastric adenocarcinoma | |||
NCT03028766 | I | Recruiting | AZD1775, Cisplatin, IMRT | A. AZD1775 + Cisplatin | AZD1775 by mouth, twice a day for 3 days on days 2, 9, 23, and 30. Cisplatin 40 mg/m2 IV delivered over 1 hour on days 2, 9, 16, 23, and 30. IMRT will be delivered 5 days a week (once daily, Monday to Friday) for 6 weeks commencing within 3 months of surgery. | Head and neck cancer | 7 | ||
B. AZD1775 + Cisplatin + IMRT | |||||||||
NCT01922076 | I | Active, not recruiting | AZD1775, RT | A. AZD1775 + RT | RT 5 days a week for 6 weeks (up to 30 fractions). Patients also receive AZD1775 orally (PO) on days 1–5 of weeks 1, 3, and 5; days 1–5 of weeks 1, 3, and 5 AND days 1, 3, and 5 of weeks 2, 4, and 6; OR days 1–5 of weeks 1–6 depending on dose level assignment. | Anaplastic astrocytoma, glioblastoma, gliosarcoma, diffuse midline glioma with histone H3 K27M mutation | Change in p-CDC2, p-HH3 and γH2AX expression | ||
NCT02095132 | I/II | Active, not recruiting | AZD1775, Irinotecan | A. Irinotecan Hydrochloride + AZD1775 | Patients receive irinotecan hydrochloride PO and AZD1775 PO on days 1–5. | Relapsed or refractory solid tumors | Changes p-CDK1 level, MYC and MYCN expression, p-WEE1 levels, EZH2 and γH2AX histone levels in tumor for correlation analyses | ||
NCT03345784 | I | Recruiting | AZD1775, Cisplatin, RT | A. AZD1775 + Cisplatin + RT | Patients undergo external beam radiation therapy on days 1–5 and receive adavosertib orally (PO) on days 1, 3, and 5 or once daily (QD) on days 1–5 and cisplatin IV over 1 hour on day 1 or 3. Cycles repeat each week for up to 5 weeks. | Cervical, vaginal, or uterine cancer | (p)CDC2, Ki67, γH2AX, pH3, and CC3. | ||
NCT01849146 | I | Active, not recruiting | AZD1775, Temozolomide, RT | A. AZD1775 + Temozolomide + RT | A. Patients receive adavosertib orally (PO) on days 1, 3, and 5 or days 1–5 weekly and temozolomide PO once daily (QD) for 6 weeks. Patients also undergo concurrent RT 5 days per week for 6 weeks. | Newly diagnosed or recurrent glioblastoma | pRb (S807/811), Ki67, p-CDC2, P-gp, cleaved caspase (apoposis), pWee1 expression levels, TP53 mutation status, MGMT methylation | 8 | |
B. AZD1775 + Temozolomide | B. Patients receive adavosertib PO QD on days 1, 3, and 5 or 1–5 weekly, and temozolomide PO QD on days 1–5. Treatment repeats every 28 days for up to 6 cycles. | ||||||||
NCT02272790 | II | Active, not recruiting | AZD1775, Gemcitabine, Paclitaxel, and others | A. AZD1775 + Gemcitabine | A. AZD1775 (175 mg PO) will be taken on Days 1–2, 8–9, and 15–16. Gemcitabine 800 mg/m² will be administered IV on days 1, 8, and 15 of each 28 day cycle. | TP53-mutated | mPFS (Arm C): 10.1 months | 9 | |
B. AZD1775 + Paclitaxel | B. Five doses of AZD1775 (225 mg PO BID) will be taken in approximate 12 hour intervals over 2.5 days weekly (Days 1–3, 8–10, and 15–17). Weekly paclitaxel 80 mg/m² IV will be administered on Day 1, 8, and 15 of each 28 day cycle. | Platinum-Resistant Epithelial Ovarian, Fallopian Tube, or Primary Peritoneal Cancer | ORR: 1/9 (A), 11/38 (B), 7/23 (C), 8/12 (C2) | ||||||
C/C2. AZD1775 + Carboplatin | C. Adavosertib 225 mg orally BID (5 doses over 3 days) on Days 1–3 of 21 day cycles. Carboplatin AUC 5 IV on Day 1 of 21 day cycles. | mPFS: 1.7. (A), 5.5 (B), 4.2 (C), and 12.0 (C2) months | |||||||
C2. Adavosertib 225 mg orally BID (5 doses over 3 days) on Days 1–3, 8–10, and 15–17 of 21 day cycles. Carboplatin AUC 5 IV on Day 1 of 21 day cycles. | |||||||||
NCT02937818 | II | Active, not recruiting | AZD1775, Carboplatin and others | A. AZD1775 + Carboplatin | AZD1775 twice daily (oral) for 2.5 days from Day 1 + CBDP area under the curve 5 (Day 1) | Platinum Refractory Extensive-Stage Small-Cell Lung Cancer | |||
NCT01164995 | II | Unknown | AZD1775, Carboplatin | A. AZD1775 + Carboplatin | Carboplatin AUC5 (IV. 30 min) at day 1. Concomitantly with the start of the carboplatin infusion 225 mg of AZD1775 will be administered as an oral capsule, followed by 4 additional doses at 12 hour increments (= 5 BID doses of AZD1775 in 2.5 days in total). | TP53-mutated Epithelial Ovarian Cancer | ORR: 43% | p-CDC2 levels on Day 1 and Day 3 post-AZD1775 | 10 |
mPFS: 5.3 months | |||||||||
OS: 12.6 months | |||||||||
NCT02341456 | Ib | Completed | AZD1775, Paclitaxel, Carboplatin | A. AZD1775 + Paclitaxel + Carboplatin | AZD1775 will be administered orally as a single dose on Day 1 Cycle 0. Following a 5±2 days washout period, AZD1775 (5 doses BID over 2.5 days) will be taken in combination with paclitaxel and carboplatin in each 21-day cycle for 6 cycles. Following 6 cycles of combination treatment, patients may continue on AZD1775 monotherapy (5 doses BID Day 1 to Day 2.5 in each 21-day cycle) at the investigator's discretion. | Advanced Solid Tumours | PR: 16.7% (2/12) | 11 | |
NCT02037230 | I/II | Completed | AZD1775, Gemcitabine, Radiotherapy | A. AZD1775 + Gemcitabine + Radiation Therapy | AZD1775 will be given as an oral capsule on days 1 and 2, and on days 8 and 9. Gemcitabine 1000 mg/m2 will be infused over 30 minutes on days 1 and 8. 52.5 Gy in 25 fractions (2.1 Gy/fraction), using intensity modulated radiation therapy (IMRT). Radiation therapy will be administered after chemotherapy. | Unresectable Adenocarcinoma of the Pancreas | mPFS: 9.4 months | p-CDC2 (IHC) | 12 |
OS: 21.7 months | |||||||||
NCT01357161 | II | Completed | AZD1775, Paclitaxel, Carboplatin | A. AZD1775 225 mg + Paclitaxel + Carboplatin | A. 225 mg AZD1775 twice daily (BID) starting on Day 1 of Cycle 1 (cycle = 21 days) for a total of 5 doses. Participants receive AZD1775 in combination with paclitaxel (175 mg/m2) and carboplatin (area under the curve [AUC] 5). (Arm B: Placebo instead of AZD1775) | TP53-mutated non-low grade, non-borderline ovarian, fallopian tube, or primary peritoneal cancer which has progressed after paclitaxel/platinum-based therapy | mPFS: 7.9 (A) vs. 7.3 months (B) | TP53 mutation subtypes | 13, 30 |
B. Placebo + Paclitaxel + Carboplatin | (HR = 0.63, P = 0.080 | ||||||||
ORR: 1.4% vs. 75.8% (P = 0.459) | |||||||||
NCT02513563 | II | Recruiting | AZD1775, Carboplatin, Paclitaxel | A. AZD1775 + Carboplatin + Paclitaxel | IV carboplatin AUC 5, IV paclitaxel 175 mg/m2, and oral AZD1775 225 mg twice/day for 2.5 days every 21 days for 4–6 cycles followed by maintenance AZD1775 at the same doses | Squamous Cell Lung Cancer | PR: 30% (3/10) | Correlative analyses of p53, PAXIP1, and WEE1 | 14 |
SD: 50% (5/10) | |||||||||
NCT02508246 | I | Completed | AZD1775, Cisplatin, Docetaxel | A. AZD1775 + Cisplatin + Docetaxel | WEE1 inhibitor AZD1775 orally (PO) twice daily (BID) on days 2–4, 9–11, and 16–18, and day -7 prior to course 1, day 1 for PD assessment. Patients also receive cisplatin IV on days 1 (or up to two days after last dose of WEE1 inhibitor AZD1775 lead-in is completed), 8 (or 7 days after first chemotherapy dose), and 15, and docetaxel IV on days 1, 8, and 15. | Squamous Cell Carcinoma of the Head and Neck (HNSCC) | CR: 2/10 | TP53 mutation status, HPV status, WEE1 and p-CDC2 levels | 15 |
PR: 5/10 | |||||||||
NCT02101775 | II | Active, not recruiting | AZD1775, Gemcitabine Hydrochloride | A. AZD1775 + Gemcitabine Hydrochloride | A. Patients receive WEE1 inhibitor AZD1775 orally (PO) on days 1, 2, 8, 9, 15, and 16 and gemcitabine hydrochloride intravenously (IV) over 30 minutes on days 1, 8, and 15. Courses repeat every 28 day. (Arm B: Placebo instead of AZD1775) | Recurrent Ovarian, Primary Peritoneal, or Fallopian Tube Cancer | pCDC2 and γH2AX levels, TP53 mutations | ||
B. Placebo + Gemcitabine Hydrochloride | |||||||||
NCT00648648 | I | Completed | AZD1775, Cisplatin, Carboplatin, Gemcitabine | A. AZD1775 + Gemcitabine | 1) AZD1775 +Gemcitabine (1,000 mg/m2), 2) AZD1775 + Cisplatin (75 mg/m2), or 3) AZD1775 +Carboplatin at an area under the time curve concentration of 5 mg/min/mL (AUC5). Following completion of Part 2-A, AZD1775 will be administered twice daily (BID) for 2.5 days (multi-dose) starting concomitantly with each administration of chemotherapy in Part 2-B. | Advanced solid tumors | SD: 53% (94/176) | pCDC2 24 and 48 hours post-AZD1775 | 16 |
B. AZD1775 + Cisplatin | PR: 10% (17/176) | ||||||||
C. AZD1775 + Carboplatin | |||||||||
NCT03012477 | II | Active, not recruiting | Cisplatin | A. Cisplatin + AZD1775 | Cisplatin will be given on the first day of every cycle. AZD1775 will be administered in clinic on the first day of the second cycle. Subsequent doses will be taken approximately 12 hours apart for a total of five doses. | Breast cancer | |||
NCT02585973 | Ib | Active, not recruiting | AZD1775, Cisplatin, IMRT | A. AZD1775 + Cisplatin + IMRT | AZD1775 given twice daily (BID) for three consecutive days (M-W) concomitantly with standard of care cisplatin (40 mg/m2 IV infused over 1-hour D1 of each week of radiation) and radiation (Total dose will be 70 Gy at 2 Gy/fx, 35 fractions, Mon to Fri, for 7 weeks) | HNSCC of the oropharynx, larynx, hypopharynx, or oral cavity | p-CDC2 and γH2AX levels, TP53 mutation analysis | ||
NCT02797977 | I/II | Active, not recruiting | SRA737, Gemcitabine, Cisplatin | A. SRA737 + Gemcitabine + Cisplatin | SRA737 will be administered orally on Days 2, 3, 9, 10, 16, and 17 of each 28-day cycle. Subjects will receive a single dose of SRA737 between 4 to 7 days prior to starting the first cycle. Gemcitabine will be administered intravenously on Days 1, 8, and 15 of each 28-day cycle. | HGSOC, SCLC, STS, and cervical/anogenital cancer | |||
NCT04023669 | I | Recruiting | Prexasertib (LY2606368), Gemcitabine, Cyclophosphamide | A. Prexasertib + Cyclophosphamide | A. Cyclophosphamide IV on days 1 and 15 and prexasertib IV on days 2 and 16. Cycles repeat every 28 days for up to 24 months (26 cycles). | Medulloblastoma | |||
B. Prexasertib + Gemcitabine | B. Gemcitabine IV on days 1 and 15 and prexasertib IV on days 2 and 16. Cycles repeat every 28 days for up to 24 months (26 cycles). | ||||||||
NCT01139775 | I/II | Completed | Pemetrexed, Prexasertib (LY2606368), Cisplatin | A. Pemetrexed + Prexasertib + Cisplatin | Pemetrexed 500 milligrams per meter square (mg/m2) + cisplatin 75 mg/m2 on Day 1 (Arm A: LY2603618 at 130–275 mg on Day 2) | Nonsquamous NSCLC | mPFS: 4.7 (A) vs. 1.5 months (B (P = 0.022) | 17, 18 | |
B. Pemetrexed + Cisplatin | |||||||||
NCT01870596 | II | Completed | SCH 900776, Cytarabine | A. SCH 900776 + Cytarabine | Patients receive cytarabine IV continuously over 72 hours on days 1–3 and 10–12 and Chk1 inhibitor SCH 900776 IV over 30 minutes on days 2, 3, 11, and 12. | Acute Myeloid Leukemia | CR: 36% (5/14) (A) vs. 44% (8/18) (B) | 19 | |
B. Cytarabine | PR: 7% (1/14) vs. 6% (1/14) (B) | ||||||||
mPFS: 5.9 vs. 4.1 months, p = ns) | |||||||||
NCT02649764 | I | Active, not recruiting | Cytarabine, Prexasertib (LY2606368), Fludarabine Phosphate | A. Cytarabine + Prexasertib + Fludarabine Phosphate | Patients ≤65 years of age: receive fludarabine IV over approximately 2 hours on days 1–4, cytarabine IV over 4 hours on days 1–4, and prexasertib (LY2606368) IV over approximately 2 hours on days 1, 3, and 4 or on days 1–4. Patients > 65 years of age: receive fludarabine IV over approximately 2 hours on days 1–3, cytarabine IV over 4 hours on days 1–3, and prexasertib (LY2606368) IV over approximately 2 hours on days 1, 3, and 4 or on days 1–4. Treatment for both age groups repeats every 28 days for up to 5 courses. | Relapsed or refractory (first or second salvage) AML or high-risk myelodysplastic syndrome (HRMDS). | Phosphorylated H2AX, Chk1/2, Cdc25, Rb, Akt, and CDC2 levels (WB or Flow Cytometry) | ||
NCT02124148 | I | Completed | Prexasertib (LY2606368), Cisplatin, Pemetrexed, Fluorouracil, and others | A/A2/A3. Prexasertib + Cisplatin | A. Prexasertib and cisplatin administered IV once every 21 days. | Advanced Cancer | CR: 0% (A), 0% (A2), 5% (1/19) (A3) | 20 | |
C. Prexasertib + Pemetrexed | A2: Prexasertib and cisplatin administered IV every 21 days; G-CSF administered subcutaneously (SC) starting approximately 24 hours after each prexasertib dose every 21 days. | Part B: KRAS wild-type colorectal cancer | PR: 21% (3/9) (A), 0% (A2), 11% (2/19) (A3) | ||||||
D. Leucovorin + 5-FU + Prexasertib | Part A3: Cisplatin administered IV on day one and prexasertib administered IV on day two once every 21 days. | SD: 14% (2/9) (A), 50% (12/24) (A2), 37% (7/19) (A3) | |||||||
Part C: Pemetrexed administered IV on day one and prexasertib administered IV on day one and two every 21 days. | |||||||||
Part D: Leucovorin administered IV on day one, 5-FU administered IV bolus on day one and by continuous IV on days one to three (46 hours), and prexasertib administered IV on day three every 14 days | |||||||||
NCT01341457 | I | Completed | LY2603618, Gemcitabine | A. 170 mg LY2603618 + Gemcitabine | Gemcitabine 1,000 mg/m2 IV on days 1, 8, and 15. 170 or 230 mg LY2603618 administered intravenously on days 2, 9, and 16 of at least one 28-day cycle. | Solid advanced or metastatic tumors | Best OR: 2/7 (A), 5/10 (B) | ||
B. 230 mg LY2603618 + Gemcitabine | |||||||||
NCT00839332 | I/II | Completed | LY2603618, Gemcitabine | A. LY2603618, Gemcitabine | 1,000 mg/m2 gemcitabine as a 30-minute continuous IV infusion once per week 24 hours prior to LY2603618. | Pancreatic cancer | OS: 7.8 (A) vs. 8.3 months (B) | 21 | |
B. Gemcitabine | Phase II: 230 mg LY2603618 as a 1-hour continuous IV infusion once per week for 3 weeks, followed by 1 week of rest. | ||||||||
NCT00988858 | II | Completed | LY2603618, Pemetrexed | 500 mg/m2 Pemetrexed IV on Day 1 and 150 mg/m2 IV on day 2 | Advanced or metastatic NSCLC | PR: 9.1% (5/55) | 22 | ||
SD: 36.% (20/50) | |||||||||
mPFS: 2.3 months | |||||||||
NCT04095221 | I/II | Recruiting | Prexasertib (LY2606368), Irinotecan | A. Prexasertib + Irinotecan | 3 + 3 design | Relapsed or Refractory Desmoplastic Small Round Cell Tumor and Rhabdomyosarcoma | |||
NCT02555644 | I | Completed | Prexasertib (LY2606368), Cisplatin, Cetuximab, IMRT | A. Prexasertib + Cisplatin + Radiation Therapy | A. Prexasertib administered IV every 14 days. Cisplatin administered IV every 7 days. IMRT administered 5 days per week. | HNSCC of the oropharynx, hypopharynx, or larynx, or SCC of the anus | |||
B. Prexasertib + Cetuximab + Radiation Therapy | B. Prexasertib administered IV every 14 days. Cetuximab administered IV every 7 days. IMRT administered 5 days per week. | ||||||||
NCT00779584 | I | Completed | MK-8776, (SCH 900776) | A. AZD1775 | IV infusion with MK-8776 at seven dose levels ranging from 10 to 150 mg/m2 as monotherapy and then in combination with gemcitabine 800 mg/m2 (part A, n = 26) or gemcitabine 1,000 mg/m2 (part B, n = 17). | Advanced solid tumor malignancy or lymphoma | PR: 7% (2/30) | 23 | |
Gemcitabine | B. AZD1775 + Gemcitabine | SD: 43% (13/30) (A) | |||||||
NCT00045513 | I/II | Completed | UCN-01, Fludarabine Phosphate | A. UCN-01 + Fludarabine Phosphate | Patients receive UCN-01 IV over 3 hours on day 1 and fludarabine IV over 30–60 minutes on days 1–5. | Chronic Lymphocytic Leukemia or Lymphocytic Lymphoma | |||
NCT00006464 | I | Completed | UCN-01, Cisplatin | A. Cisplatin + UCN-01 | Patients receive cisplatin IV over 1 hour on day 1 and UCN-01 IV continuously over 36–72 hours beginning on day 2. | Advanced or metastatic solid tumor incurable by surgery or other standard therapy | |||
NCT00700336 | I/II | Completed | Pemetrexed, Cisplatin and CBP501 | A. Pemetrexed + Cisplatin + CBP501 | Advanced solid tumors and in chemotherapy-naïve patients with malignant pleural mesothelioma | ||||
B. Pemetrexed + Cisplatin | |||||||||
NCT00551512 | I | Completed | Cisplatin, CBP501 | A. CBP501 | CBP501 alone (D1/D8/D15, from 0.9 mg/m2), or with Cisplatin (both on D1, from 3.6 mg/m2 CBP501, 50 mg/m2 cisplatin). | Advanced refractory solid tumors (Arm B expansion: synchronous ovarian and endometrial carcinoma) | PR: 0/30 (A), 2/14 (B) | Rad51, BRCA1, ATM, FANCD2 and MUS81, XPF and Polη, ATM and phospho-MAPKAP-K2 levels (IHC) | 24 |
B. Cisplatin + CBP501 | SD: 7/30 (A), 5/14 (B) | ||||||||
NCT00413686 | I | Completed | AZD7762, Gemcitabine | In the first cycle (cycle 0), patients received a single dose of AZD7762 administered as a 60-minutes IV infusion on days 1 and 8 of a 14-day run-in cycle. In subsequent cycles, patients received AZD7762 at the same dose as cycle 0 in combination with 750 or 1,000 mg/m2 gemcitabine administered as a 30-minute IV infusion. The first seven patients received combination treatment on days 1, 8, and 15 of a 28-day cycle. | Advanced Solid Malignancies | PR: 1/38 (AZD7762 6 mg/gemcitabine 750 mg/m2 and AZD7762 9 mg cohort) | 25 | ||
SD: 5/38 (AZD7762 6 mg/gemcitabine 750 mg/m2, AZD7762 14 mg and AZD7762 30 mg cohorts, n = 1 patient per cohort, and AZD7762 40 mg cohort n = 2 patients) | |||||||||
NCT00031681 | I | Completed | UCN-01, Irinotecan Hydrochloride | A. UCN-01 + Irinotecan Hydrochloride | Patients receive irinotecan hydrochloride intravenously (IV) over 90 minutes on days 1, 8, 15, and 22 and 7-hydroxystaurosporine IV over 3 hours on days 2 and 23. | Metastatic or Unresectable Solid Tumors or Triple Negative Breast cancer | PR: 2/25 | TP53 mutation, p-S6, p-CDC2, p-Chk1(S317), p-HH3, γH2AX and cleaved caspase 3 (IHC) | 26 |
SD: 12/25 | |||||||||
NCT00036777 | I | Completed | UCN-01, Carboplatin | A. UCN-01 + Carboplatin | Patients receive carboplatin IV over 1 hour followed by UCN-01 IV over 3 hours on day 1. | Advanced solid tumors | |||
NCT00004263 | I | Completed | UCN-01, Cytarabine | A. UCN-01 + Cytarabine | Patients receive cytarabine IV over 24 hours on days 1–4 of each course. Patients receive UCN-01 IV over 24 hours on days 2–4 of course 1 and over 36 hours beginning on day 2 of subsequent courses. | Refractory or relapsed acute myelogenous leukemia or myelodysplastic syndrome | SD: 2/11 | Akt, p-Akt(S473), and p38α | 27 |
MAPK levels (IHC) | |||||||||
NCT00039403 | I | Completed | UCN-01, Gemcitabine Hydrochloride | A. UCN-01 + Gemcitabine Hydrochloride | Patients receive gemcitabine IV over 1–2 hours on days 1 and 8 followed by UCN-01 IV over 3 hours on day 1. | Adenocarcinoma of the pancreas | |||
NCT00098956 | II | Completed | UCN-01, Topotecan | A. UCN-01 + Topotecan | Patients receive topotecan IV over 30 minutes on days 1–5 and UCN-01 IV over 3 hours on day 1. | Relapsed or progressed SCLC | |||
NCT00045747 | II | Completed | UCN-01, Fluorouracil | A. UCN-01 + Fluorouracil | Patients receive fluorouracil IV over 24 hours on days 1, 8, 15, and 22. Patients also receive UCN-01 IV continuously over 72 hours (course 1 only) beginning on day 2. In subsequent courses, UCN-01 is infused over 36 hours. | Gemcitabine-refractory metastatic pancreatic cancer | |||
NCT00045500 | I | Completed | UCN-01, Prednisone | A. UCN-01 + Prednisone | Patients receive oral prednisone daily on days 1–5 and UCN-01 IV over 36–72 hours on days 3–5. | Refractory solid tumors or lymphomas | |||
NCT00047242 | I | Completed | UCN-01, Irinotecan Hydrochloride | A. UCN-01 + Irinotecan Hydrochloride | Patients receive UCN-01 IV over 3 hours on day 1 and irinotecan IV over 90 minutes on days 1 and 8 | Solid tumors | SD: 4/16 | 28 | |
NCT00042861 | I | Completed | UCN-01, Fluorouracil | A. UCN-01 + Fluorouracil | Patients receive leucovorin calcium (CF) IV over 2 hours and 5-FU IV (at the midpoint of CF administration) on day 1, followed by UCN-01 IV over 36–72 hours, on weeks 1–3. | Metastatic or unresectable solid tumors | |||
NCT00072267 | II | Completed | UCN-01, Topotecan Hydrochloride | A. UCN-01 + Topotecan Hydrochloride | Patients receive UCN-01 IV over 3 hours on day 1 and topotecan IV over 30 minutes on days 1–5. | Recurrent, persistent, or progressive advanced ovarian epithelial, primary peritoneal, or fallopian tube cancer | |||
NCT00019838 | I | Completed | UCN-01, Fludarabine Phosphate | A. UCN-01 + Fludarabine Phosphate | Patients receive UCN-01 IV over 72 hours on days 1–3 alone during course 1 and over 36 hours on days 1–2 during courses 2–7. Patients also receive fludarabine IV over 30 minutes beginning on day 1 and continuing for up to 5 days during courses 2–7. | Leukemia | OR: 38% | Apoptosis | 29 |
Lymphoma | CR: 1/18 | ||||||||
PR: 6/18 | |||||||||
SD: 7/18 | |||||||||
NCT00004059 | I | Completed | UCN-01, Fluorouracil | A. UCN-01 + Fluorouracil | Patients receive fluorouracil IV over 24 hours on days 1, 8, 15, and 22. Patients receive an initial dose of UCN-01 IV over 72 hours beginning on day 2 during course 1 and then maintenance UCN-01 IV over 36 hours beginning on day 2 during subsequent courses. | Advanced or refractory solid tumors | |||
NCT00045175 | I | Completed | UCN-01, Topotecan Hydrochloride | A. UCN-01 + Topotecan Hydrochloride | Patients receive UCN-01 IV over 3 hours on day 1 and topotecan IV over 30 minutes on days 1–5. | Fallopian tube cancer | |||
Ovarian cancer | |||||||||
Primary peritoneal cavity cancer |
Clinical trials of cell-cycle checkpoint kinase inhibitors (CHK1/2, WEE1, ATR) in combination with chemotherapy agents or radiotherapy. Search terms were WEE1, ATM, ATR, CHK1, and CHK2, as well as the known drug names, with “combination”. Only interventional trials that are recruiting, active, or completed were included in the table, not those that were terminated. All treatment schedules are 21-day cycles in the absence of disease progression or unacceptable toxicity unless indicated otherwise.
Abbreviations: AML, acute myelogenous leukemia; b.i.d., twice daily; CDC2, cyclin dependent kinase 1; CLL, chronic lymphocytic leukemia; CR, complete response; ctDNA, circulating tumor DNA; HNSCC; head and neck small cell carcinoma; IV, intravenous administration; OS, overall survival; P, P value of two-sided statistical test; PFS, progression-free survival; p.o., oral administration; PR, partial response; q.i.d., once daily; QD; once daily; RR, overall response rate (complete and partial responses); SCLC; small cell lung carcinoma; SD, stable disease; TNBC; triple-negative breast cancer; vs., versus; WBRT; whole-brain radiotherapy.
References
1Telli, M. et al. Abstract OT2–07–07: ATR inhibitor M6620 (formerly VX-970) with cisplatin in metastatic triple-negative breast cancer: Preliminary results from a phase 1 dose expansion cohort (NCT02157792). in (2018). doi:10.1158/1538–7445.sabcs17-ot2–07–07.
2Thomas, A. et al. Phase I study of ATR inhibitor M6620 in combination with topotecan in patients with advanced solid tumors. J. Clin. Oncol. (2018). doi:10.1200/JCO.2017.76.6915.
3Konstantinopoulos, P. A. et al. Berzosertib plus gemcitabine versus gemcitabine alone in platinum-resistant high-grade serous ovarian cancer: a multicentre, open-label, randomised, phase II trial. Lancet Oncol. (2020). doi:10.1016/S1470–2045(20)30180–7.
4O'Sullivan Coyne, G. H. et al. Phase I trial of the triplet M6620 (formerly VX970) + veliparib + cisplatin in patients with advanced solid tumors. J. Clin. Oncol. (2018). doi:10.1200/jco.2018.36.15_suppl.2549.
5Dillon, M. T. et al. PATRIOT: A phase I study to assess the tolerability, safety and biological effects of a specific ataxia telangiectasia and Rad3-related (ATR) inhibitor (AZD6738) as a single agent and in combination with palliative radiation therapy in patients with solid tumours. Clin. Transl. Radiat. Oncol. (2018). doi:10.1016/j.ctro.2018.06.001.
6Lee, J. et al. Results from a phase I, open-label study of ceralasertib (AZD6738), a novel DNA damage repair agent, in combination with weekly paclitaxel in refractory cancer (NCT02630199). J. Clin. Oncol. (2020). doi:10.1200/jco.2020.38.15_suppl.3503.
7Kong, A. et al. Phase I trial of WEE1 inhibition with chemotherapy and radiotherapy as adjuvant treatment, and a window of opportunity trial with cisplatin in patients with head and neck cancer: The WISTERIA trial protocol. BMJ Open (2020). doi:10.1136/bmjopen-2019–033009.
8Alexander, B. M. et al. Phase I study of AZD1775 with radiation therapy (RT) and temozolomide (TMZ) in patients with newly diagnosed glioblastoma (GBM) and evaluation of intratumoral drug distribution (IDD) in patients with recurrent GBM. J. Clin. Oncol. (2017). doi:10.1200/jco.2017.35.15_suppl.2005.
9Moore, K. N. et al. Adavosertib with chemotherapy (CT) in patients (pts) with platinum-resistant ovarian cancer (PPROC): An open label, four-arm, phase II study. J. Clin. Oncol. (2019). doi:10.1200/jco.2019.37.15_suppl.5513.
10Leijen, S. et al. Phase II study of WEE1 inhibitor AZD1775 plus carboplatin in patientswith tp53-mutated ovarian cancer refractory or resistant to first-line therapy within 3 months. J. Clin. Oncol. (2016). doi:10.1200/JCO.2016.67.5942.
11Kato, H. et al. Safety, pharmacokinetics, and clinical activity of adavosertib in combination with chemotherapy in Asian patients with advanced solid tumors: phase Ib study. Target. Oncol. (2020). doi:10.1007/s11523–020–00701–5.
12Cuneo, K. C. et al. Dose escalation trial of the WEE1 inhibitor adavosertib (AZD1775) in combination with gemcitabine and radiation for patients with locally advanced pancreatic cancer. J. Clin. Oncol. (2019). doi:10.1200/JCO.19.00730.
13Oza, A. M. et al. An international, biomarker-directed, randomized, phase II trial of AZD1775 plus paclitaxel and carboplatin (P/C) for the treatment of women with platinum-sensitive, TP53 -mutant ovarian cancer. J. Clin. Oncol. (2015). doi:10.1200/jco.2015.33.15_suppl.5506.
14Gray, J. E. et al. Phase II trial of AZD1775 in combination with carboplatin and paclitaxel in stage IV squamous cell lung cancer (sqNSCLC): Preliminary results. J. Clin. Oncol. (2017). doi:10.1200/jco.2017.35.15_suppl.e20672.
15Mendez, E. et al. A phase I clinical trial of AZD1775 in combination with neoadjuvant weekly docetaxel and cisplatin before definitive therapy in head and neck squamous cell carcinoma. Clin. Cancer Res. (2018). doi:10.1158/1078–0432.CCR-17–3796.
16Leijen, S. et al. Phase I study evaluating WEE1 inhibitor AZD1775 as monotherapy and in combination with gemcitabine, cisplatin, or carboplatin in patients with advanced solid tumors. J. Clin. Oncol. (2016). doi:10.1200/JCO.2016.67.5991.
17Wehler, T. et al. A randomized, phase 2 evaluation of the CHK1 inhibitor, LY2603618, administered in combination with pemetrexed and cisplatin in patients with advanced nonsquamous non‐small cell lung cancer. Lung Cancer (2017). doi:10.1016/j.lungcan.2017.03.001.
18Calvo, E. et al. Preclinical analyses and phase I evaluation of LY2603618 administered in combination with pemetrexed and cisplatin in patients with advanced cancer. Investigational New Drugs (2014). doi:10.1007/s10637–014–0114–5.
19Webster, J. A. et al. Randomized phase II trial of cytosine arabinoside with and without the CHK1 inhibitor MK-8776 in relapsed and refractory acute myeloid leukemia. Leuk. Res. (2017). doi:10.1016/j.leukres.2017.09.005.
20Patel, M. R. et al. A phase 1b dose-escalation study of prexasertib, a checkpoint kinase 1 (CHK1) inhibitor, in combination with cisplatin in patients with advanced cancer. J. Clin. Oncol. (2018). doi:10.1200/jco.2018.36.15_suppl.2579.
21Laquente, B. et al. A phase II study to evaluate LY2603618 in combination with gemcitabine in pancreatic cancer patients. BMC Cancer (2017). doi:10.1186/s12885–017–3131-x.
22Scagliotti, G. et al. Phase II evaluation of LY2603618, a first-generation CHK1 inhibitor, in combination with pemetrexed in patients with advanced or metastatic non-small cell lung cancer. Invest. New Drugs (2016). doi:10.1007/s10637–016–0368–1.
23Daud, A. I. et al. Phase I dose-escalation trial of checkpoint kinase 1 inhibitor MK-8776 as monotherapy and in combination with gemcitabine in patients with advanced solid tumors. J. Clin. Oncol. (2015). doi:10.1200/JCO.2014.57.5027.
24Shapiro, G. I. et al. Phase I studies of CBP501, a G2 checkpoint abrogator, as monotherapy and in combination with cisplatin in patients with advanced solid tumors. Clin. Cancer Res. (2011). doi:10.1158/1078–0432.CCR-10–2345.
25Sausville, E. et al. Phase I dose-escalation study of AZD7762, a checkpoint kinase inhibitor, in combination with gemcitabine in US patients with advanced solid tumors. Cancer Chemother. Pharmacol. (2014). doi:10.1007/s00280–014–2380–5.
26Fracasso, P. M. et al. A Phase 1 study of UCN-01 in combination with irinotecan in patients with resistant solid tumor malignancies. Cancer Chemother. Pharmacol. (2011). doi:10.1007/s00280–010–1410–1.
27Gojo, I. et al. Phase i study of UCN-01 and perifosine in patients with relapsed and refractory acute leukemias and high-risk myelodysplastic syndrome. Invest. New Drugs (2013). doi:10.1007/s10637–013–9937–8.
28Jimeno, A. et al. Phase I and pharmacokinetic study of UCN-01 in combination with irinotecan in patients with solid tumors. Cancer Chemother. Pharmacol. (2008). doi:10.1007/s00280–007–0485–9.
29Marti, G. E. et al. Phase i trial of 7-hydroxystaurosporine and fludararbine phosphate: In vivo evidence of 7-hydroxystaurosporine induced apoptosis in chronic lymphocytic leukemia. Leuk. Lymphoma (2011). doi:10.3109/10428194.2011.589547.
30Oza, A. M., Estevez-Diz, M., Grischke, E. M., Hall, M., Marmé, F., Provencher, D. M., Uyar, D. S., Weberpals, J. I., Wenham, R. M., Laing, N., Tracy, M., Freshwater, T., Lee, M. A., Liu, J., Qiu, J., Rose, S., Rubin, E. H., & Moore, K. N. (2020). A biomarker-enriched, randomized Phase II trial of adavosertib (AZD1775) plus paclitaxel and carboplatin for women with platinum-sensitive TP53-mutant ovarian cancer. Clinical cancer research: an official journal of the American Association for Cancer Research, clincanres.0219.2020. Advance online publication. https://doi.org/10.1158/1078-0432.CCR-20-0219.
Choice and scheduling of combination therapies
As previously discussed, preclinical studies have demonstrated that the delayed administration of cell-cycle checkpoint kinase inhibitors after S-phase–specific chemotherapeutic agents is important for maximal cell killing. Even though this delayed drug schedule has been adopted in many of the clinical trials presented in Table 1, and results have been encouraging, other studies have shown limited efficacy and could have benefited from timed sequential therapy, a therapeutic strategy that exploits cell-cycle kinetics by administering a second drug in close approximation with the damage and/or replication stress induced by the first drug.
One of the most prominent examples of recent success comes from a phase II trial of gemcitabine and berzosertib, a selective ATR inhibitor, in high-grade serous ovarian cancer (NCT02595892), where it was shown that combination therapy increased progression-free survival compared with gemcitabine alone [mPFS was 22·9 weeks (17·9–72·0) for gemcitabine + ATRi and 14·7 weeks (90% confidence interval (CI) 9·7–36·7) for gemcitabine alone (HR, 0·57; 90% CI 0·33–0·98; one-sided log-rank test P = 0·044; ref. 63)]. In this study, the ATR inhibitor, berzosertib, was administered 24 hours after gemcitabine, highlighting the enhanced clinical efficacy when the optimal sequence and delayed schedule of administration of checkpoint kinase inhibitors in combination with chemotherapy is applied in the clinic. Similarly, a phase II study (NCT01357161) assessing the effectiveness of AZD1775 plus paclitaxel and carboplatin (P/C) inTP53-mutant ovarian cancer with an analogous dosing strategy (AZD1775 or placebo bid for 2.5 days plus paclitaxel (175 mg/m2; i.v.) and carboplatin (AUC5; i.v., on day 1) also found that AZD1775/P/C was associated with a significant increase in progression-free survival (PFS) when compared with P/C alone (70).
Those using concomitant treatment [gemcitabine and AZD7762 (ATRi; NCT00413686)], or a short (30 minutes) cell-cycle checkpoint inhibitor bolus 24 hours postchemotherapy [gemcitabine and CHK1i (NCT00839332)] have, on the contrary, shown very limited improved efficacy over standard of care chemotherapy. This could be due to inadequate target engagement, as the above-mentioned studies used continuous intravenous administration of the cell-cycle checkpoint kinase inhibitor (>1 hour, up to 1–3 days), or the ability of cells to recover from checkpoint inhibition. A phase I study tried to evaluate the efficacy of a 3- versus 24-hour PF-00477736 (CHK1i)-intravenous infusion 24 hours after gemcitabine treatment in Advanced Solid Tumors (NCT00437203), which would have provided an interesting insight into the importance of timing of administration and target engagement, but unfortunately, the study was terminated prematurely due to business reasons.
A phase I study combining AZD1775 (WEE1 inhibitor) and carboplatin in platinum-resistant ovarian cancer (NCT02272790), showed an objective response rate (ORR) of 67% and a median progression-free survival (mPFS) of 12.0 months when carboplatin was administered on day 1 and AZD1775 on days 1–3, 8–10, and 15–17 of 21 day cycles. When the same drugs were used in a different regimen (carboplatin was administered on day 1 and AZD1775 only on days 1–3), mPFS was 4.2 months, with an ORR of only 30%. This indicates the importance of continued target inhibition (i.e., WEE1) in the presence of DNA damage and suggests that repeated target engagement is necessary to block the ability of cells to recover from checkpoint inhibition and thus for the therapy to be effective.
Below, we consider two examples of trials that have been unsuccessful and discuss these in the context of the preclinical findings presented in the previous section. First, a phase I/II study designed to compare the OS of LY2603618/gemcitabine to gemcitabine alone in locally advanced or metastatic pancreatic cancer (NCT00839332), as well as another study testing gemcitabine and CHK1i on a similar schedule in Japanese patients (NCT00937664, terminated), did not only reveal a higher number of serious adverse effects, but also failed to observe any objective responses with concomitant administration (71). Preclinical studies have shown that concurrent CHK1 inhibition and gemcitabine failed to activate DNA helicases, which function to unwind the DNA in the absence of nucleotides, ultimately resulting in DNA cleavage and replication catastrophe (29, 46). These two clinical studies demonstrate that sensitization of this drug combination in patients does not occur with concurrent treatment, which aligns with the preclinical findings.
A final example is that of AZD1775 (WEE1i) and gemcitabine (NCT02272790) in TP53-mutated epithelial ovarian cancer, where AZD1775 was administered orally on days 1 and 2 and gemcitabine intravenously on day 1 (week 1–3). The mPFS in this group was 1.7 months, versus 5.5 months (when AZD1775 was combined with paclitaxel), and 12.0 months (when AZD1775 was combined with carboplatin; ref. 72). The current consensus is that WEE1 does not play a direct role in replication fork protection, but rather that its effects are mediated more via its roles in replication dynamics and indirect DNA DSB generation (14). This emphasizes the need to select the agent that gives a maximally cytotoxic effect when used in combination with WEE1 inhibitors, in this case carboplatin or paclitaxel over gemcitabine, and the need to test this in preclinical models or early-phase clinical trials before starting a phase II study.
Dosage
A high number of clinical studies had to be terminated due to serious adverse effects (SAE). In most cases, toxicity is dose-dependent, as with a phase I dose-escalation study of cisplatin and UCN-01 (CHK1i), where cisplatin was infused over 1 hour before UCN-01 (45 mg/m2/d) given as a 72-hour continuous infusion. Escalation of cisplatin was planned through five dose levels at 20, 30, 45, 60, and 75 mg/m2, where accrual was halted at dose level 2 due to dose-limiting toxicities consisting of grade 5 sepsis with respiratory failure (73). A number of other examples can be found, where after a planned safety analysis of the first-line trial, dose and schedule were reduced and modified, respectively, to reduce toxicity (NCT02087241, carboplatin/pemetrexed + AZD1775 and NCT02087176, docetaxel + AZD1775).
Biomarkers and patient stratification
Molecular diagnosis and treatment stratification are at the heart of precision medicine in cancer. While the increasing availability of genomic and proteomic data has led to some key advances, molecular stratification becomes much more complex when targeting intracellular processes with multiple control mechanisms and feedback loops. Discovery and validation of cell-cycle (arrest) biomarkers has been challenging, especially as distinct predictive genes emerge from different treatment cohorts and cancer subtypes. Dysregulation of cell-cycle control can be assessed in multiple ways, but the most commonly used predictive biomarker for G1-S checkpoint deficiency is TP53 mutation status.
A phase I study evaluated AZD1775 (WEE1i) as monotherapy or in combination with chemotherapy in patients with refractory solid tumors (NCT00648648), and they correlated the response with TP53 status (74). Their study suggested that tumors from responding patients were mildly enriched for TP53 mutations, given the response rates of 21% and 12% in TP53-mutated and TP53 wild-type patients, respectively (74). The modest difference in response rate between TP53 wild-type and TP53-mutant patients was not sufficiently strong to promote the use of TP53 as a biomarker for selection of patients for Wee1 inhibitor therapy, but multiple clinical trials were, however, built on this premise. NCT00031681, NCT02157792, NCT03641313, NCT03641547, NCT02906059, NCT02448329, NCT01849146, NCT02272790, NCT01164995, NCT02087176, NCT01357161, NCT02508246, NCT02101775, NCT02585973 and other studies have subsequently implemented patient stratification (based on TP53 mutation or others) and/or retrospective mutation correlation analysis in their clinical trial design.
Some improvement on single gene (i.e., TP53) testing has been implemented. Recently, a clinical trial was launched to study AZD1775 and other drugs (as a monotherapy, nonetheless), based on a biomarker-driven treatment strategy (NCT02688894), in patients with small-cell lung cancer where patients were allocated to different treatment arms depending on the presence or absence underlying mutations in cell-cycle–related genes (CDNK2 and TP53) and the MYC family genes (75). Similarly, the FOCUS4 programme (ISRCTN90061546), consisting of multiple biomarker-stratified randomized trials, enrolls patients in different arms of the trial, depending on their molecular background. Those with TP53 + RAS mutations, a selection marker for loss of G1–S control and oncogene-driven replication stress, receive AZD1775 as a monotherapy (76). Furthermore, recent studies have suggested that more functional biomarkers of replication stress, including surrogate read-outs of the presence of ssDNA (e.g., phospho-RPA), could indicate a higher dependency on the ATR/CHK1/WEE1 pathway of the replication stress response (77). Recently, a novel transcriptomic signature of replication stress was developed, which could predict response to both ATR (P < 0.018) and WEE1 inhibitor (P < 0.029) treatment in both cell lines and human pancreatic cancer organoids. This signature takes into account expression of numerous genes involved in cell-cycle control and DNA maintenance (e.g., MYC, CCNE1, E2F1, KRAS, and CCND2) and has potential as a putative biomarker in a clinical setting (64).
Despite the limited observed clinical efficacy of WEE1 and ATR inhibitors as monotherapies in some genetic backgrounds, there might be patients, especially those with DDR-defects, that do show a partial response to monotherapy checkpoint kinase inhibitor treatment (78). For instance, the ATR inhibitor BAY 1895344 showed stronger antitumor activity in patients with solid tumors that harbor ATM loss (78). Similarly, CRISPR screens and bioinformatic analyses have recently been performed to determine which genetic alterations render cancer cells significantly more sensitive or resistant to checkpoint kinase inhibitors as a monotherapy (64, 79). Such screening has been performed for ATRi in a TP53-mutant background providing insight into which genetic background will show increased sensitivity or resistance and it is anticipated that many such screens will follow (79). However, a recent study in pancreatic cancer patient–derived cell lines shows that deficiency in DDR pathways only renders sensitivity to checkpoint kinase inhibitors when cells also suffer from intrinsic replication stress (64). Therefore, tumors that are DDR-deficient may benefit more from treatment with cytotoxic agents, such as platinum-based therapies, or PARP inhibitors, which can be combined with ATR or WEE1 inhibition if concurrent high replication stress exists within the tumor (64). These studies demonstrate the challenges that come with using checkpoint kinase inhibitors as a monotherapy.
Finally, novel biomarker approaches using other cell-cycle inhibitor(s) or combinations should be further investigated. We note that to improve the monitoring of patient responses, the development of a broad spectrum of blood-based biomarkers, such as those described here, will also be important in order to avoid further patient biopsies where possible.
Concluding Remarks and Therapeutic Outlook
Cyclotherapy approaches attempt to optimize drug selectivity against tumor cells with defective G1–S control, and this proves to be a successful therapeutic strategy if dosage and timing strategies are carefully considered in the design of clinical studies. We are suggesting that the most important consideration in combination therapy is cell-cycle specificity of the cytotoxic agent and the checkpoint kinase inhibitor. This may mean that novel combinations of cytotoxic agents with checkpoint kinase inhibitors may be more fruitful than previously approved cytotoxic agents. A number of genetic alterations or signatures of elevated replication stress could be utilized to identify tumors that are predicted to display higher sensitivity to checkpoint kinase inhibitors.
There is evidence to justify delaying administration of CHK1 or WEE1 inhibitors after DNA-damaging agents to achieve a more significant decrease in tumor growth, because such a schedule would allow cancer cells to become increasingly dependent on intra-S and G2–M checkpoint signaling for HR-mediated repair of exogenously induced replication stress and DNA damage. We propose the efficacy of combination therapies and treatment schedules should be assessed in a first cycle of drug administration for routine clinical management, which would then be continued if the predicted pharmacodynamic biomarkers are observed.
Ideally, patient treatment should be stratified according to the genetic background of their cancer as different mutations will exhibit varying degrees of sensitivity to these agents compared to normal tissue. Enhancing the therapeutic window is clinically relevant as it will reduce toxicity to the healthy cells while maintaining therapeutic benefit. Patient stratification can be achieved through predictive or preclinical testing methods, or through retrospective clinical correlation methods.
In addition, retrospective analysis of patient genomes can help guide further clinical studies. To explore the effect of the mutations mentioned above on the response rate, as well as identify other genetic markers that predict sensitivity, including mutations in genes involved in DNA repair (such as BRCA1), cell-cycle regulation (including cyclins) and oncogene-induced replication stress genes (such as c-MYC and KRAS), clinical trials with increasing patient numbers should be conducted.
A final consideration returns to fundamental science, namely, the specificity of the inhibitors. The WEE1 inhibitor, AZD1775, has been shown to inhibit PLK1 and the CHK1 inhibitor, prexaseritib (LY2606368), as well as other CHK1 inhibitors, often carry varying levels of CHK2 inhibitory activity (80). This fundamental issue with the specificity of an anticancer drug highlights the necessary and crucial interdependence of preclinical and clinical scientists, as emphasized throughout this article, to ensure that the best science is applied reliably and robustly to the life-critical field of oncology.
Authors' Disclosures
T.S. Maughan reports grants, personal fees, and nonfinancial support from AstraZeneca and grants from Merck-Serono outside the submitted work, as well as grant funding to institution from Psioxus and DMEC support from Pierre Fabre. No disclosures were reported by the other authors.