2.2.1. ATR-CHK1 inhibitors
The most important function of the ATR–CHK1 signaling pathway is to stabilize replication forks, limit the number of active origins and repair of DSB and collapsed replication forks during S phase. By repressing the origins’ firing the ATR-CHK1 pathway realizes control of the cell cycle and maintains the genomic integrity in response to DNA damage and RS (
Figure 3). On the other hand, RS that is common among different types of cancers serves as a potent activator of ATR-CHK1 signaling [
91]. Thus, the ATR-CHK1 pathway is considered as an attractive target for anticancer therapy.
ATRi including berzosertib (also known as M6620, VX-970) and gartisertib (M4344, VX-803) produced by Merck Serono, ceralasertib (AZD6738) by AstraZeneca, elimusertib (BAY1895344) by Bayer, camonsertib (RP-3500) by Repare Therapeutics, and ART0380 by Artios Pharma, have demonstrated high anti-cancer activity, especially in cancers with high RS level and increased ATR-CHK1 dependency, oncogenic Ras activation,
CCNE1 or
MYC amplification [
92]. Also small molecule ATRi have been employed to improve the efficacy of DNA damage-based chemotherapy for rapid elimination of proliferating tumor cells. It was demonstrated that ATRi could improve response of cancer cells to conventional chemotherapeutic agents as studied in a rapidly growing number of clinical trials (
Table 1).
Berzosertib is a potent and ATP-competitive selective small-molecule ATRi [
93]. Berzosertib is currently in 1/2 phase clinical trials in combination with other anticancer treatments, specifically chemotherapeutic drugs. It showed single-agent activity, as well as synergistic activity in combination with cisplatin, especially in advanced cancers with ATM aberrations confirming synthetically lethal interaction between ATM deficiency and ATR inhibition during the phase 1 clinical studies[
94,
95,
96]. Berzosertib is effective in the treatment of brain metastases from NSCLC enhancing the effect of radiation [
97], in combination with topoisomerase I inhibitor topotecan in small cell lung cancer [
98], and with cisplatin in neuroendocrine tumors [
99]. Prominent results were demonstrated on chemotherapy-resistant small cell neuroendocrine cancer and high-grade serous ovarian cancer (HGSOC), which exhibit high levels of RS. Combinations of berzosertib with topoisomerase I inhibitors and gemcitabine were synergistically cytotoxic and showed durable tumor regressions and increase in progression-free survival [
98,
100,
101]. Berzosertib demonstrated synthetic lethality
in vitro and
in vivo in tumor cells with
ARID1 deletion or mutations, one of genes commonly altered in cancers [
93,
102,
103].
AZ20 is a potent and selective ATRi that belongs to sulfonylmorpholinopyrimidines. AZ20 inhibits ATR and ATR-mediated phosphorylation of Chk1 in HT29 colorectal adenocarcinoma tumor cells. AZ20 demonstrated high antiproliferative activity against different neoplasms
in vitro and
in vivo [
104,
105,
106]. The structure of AZ20 was optimized to create AZD6738 (ceralasertib) – another ATRi with improved preclinical physicochemical and pharmacokinetic characteristics [
107,
108]. Ceralasertib was active as single agent in NSCLC cell lines [
109] and potentiated the cytotoxicity of cisplatin and gemcitabine in NSCLC cells, and ATM-deficient lung cancer xenografts [
110]. Similar synergistic effects were demonstrated for pancreatic ductal adenocarcinoma (PDAC) [
111]. Ceralasertib inhibited gemcitabine-induced CHK1 activation and prevented cell-cycle arrest, leading to the strong induction of RS markers. For instance, ATR inhibition by ceralasertib promotes sensitization to cisplatin in head and neck squamous cell carcinoma (HNSCC) regardless of presence of HPV, one of the important causes of oropharyngeal infection [
112]. There is a complex interaction between HPV and DDR proteins. HPV creates aberrant DNA structures during its rapid replication thereby activating cellular RS and recruiting DDR proteins. Subsequently, activation of the ATR signaling pathway leads to the DNA repairing, facilitating viral replication and ensuring a successful viral life cycle [
113]. Thus, in
in vivo studies, combined treatment with cisplatin and ceralasertib exhibited greater anti-tumor effects, relative to either mono agent, against both HPV+ and HPV− xenograft models, including patient-derived xenograft (PDX) models [
112]. Anti-tumor activity of ceralasertib as mono agent and with cisplatin was demonstrated on HER2-positive breast cancer in vitro [
114].
Sensitivity to ceralasertib is elevated in models with increased RS such as tumors with defects in the ATM pathway or
CCNE1 amplification [
115]. Complete loss of ATM function in PDAC models is also critical for efficacy of ATRi/gemcitabine combinational treatment [
116]. Ceralasertib also showed increased activity in other models with high genomic instability, such as a
BRCA2-mutant triple-negative breast cancer (TNBC) PDX model. Moreover, ceralasertib had combinatorial efficacy with chemotherapy medications carboplatin and irinotecan and the PARP inhibitor olaparib [
115]. Ceralasertib is currently being evaluated in a 1/2 phase clinical trials and one study is in a 3 phase and is aimed to check the efficacy and safety the inhibitor in combination with durvalumab in patients with locally advanced and metastatic NSCLC after progression on prior anti-programmed death ligand 1 (anti-PD-L1) therapy and platinum-based chemotherapy (NCT05450692,
Table 1). It has been demonstrated that the DNA damage checkpoint plays a critical role in regulating PD-L1 expression [
117]. It was demonstrated that PD-L1 expression in cancer cells is upregulated in response to DSBs and requires ATR/CHK1 kinases [
118]. Anti-PD-L1 can be combined with ATR targeted drugs to improve therapeutic response to immune checkpoint blockade therapy.
Elimusertib (BAY1895344) is a ATR kinase inhibitor in 1/2 phase clinical trials that showed synergistic antitumor activity in combination with DNA damage-inducing, repair-compromising chemotherapy or radiotherapy in preclinical cancer models. Moreover, it improved antitumor efficacy of nonsteroidal androgen receptor antagonist darolutamide in hormone-dependent prostate cancer [
119]. High-risk neuroblastomas are in a group of tumors with oncogene-induced RS because of
MYCN amplification and frequent
ALK mutations. It was shown that together with ALKi elimusertib potently inhibited cell growth, and led to complete tumor regression in mice models [
120]. Elimusertib demonstrated high efficacy against aggressive uterine leiomyosarcoma harboring ATRX mutations in
in vivo models [
121] and ovarian and uterine carcinosarcoma cell lines and xenografts [
122]. Together with PARP inhibitor elimusertib potentiated antitumor activity of HER2-targeted antibody-drug conjugates in HER2-positive cancer
in vitro and in xenograft models [
123]. Treatment by elimusertib together with anti-PD-L1 resulted in high antitumor activity in a syngeneic mice model with androgen-indifferent, aggressive prostate cancer [
124]. The inhibitor is currently in 1/2 phase clinical trials (
Table 1).
Gartisertib (M4344, VX-803) is a relatively novel ATP-competitive ATRi that is currently in clinical development. Gartisertib suppressed cancer cell proliferation at concentrations similar to elimusertib and was more potent than berzosertib and ceralasertib [
125]. It, as other ATRi, is highly synergistic with a broad range of DNA-targeting anticancer agents including topoisomerase inhibitors, gemcitabine, cisplatin, and PARPi [
125,
126]. The anticancer activity of gartisertib was demonstrated in multiple cancer cell lines, patient-derived tumor organoids, and mouse xenograft models. In combination with ATM inhibitor M4076, gartisertib demonstrated high anti-tumor efficacy in PDX models of TNBC [
127]. The inhibitor is in 1/2 phase clinical trials (
Table 1).
Camonsertib (RP-3500) is a 1/2 phase clinical-stage [
128] inhibitor that demonstrated high effectiveness in preclinical models as a monotherapy and in combination with PARPi olaparib or niraparib [
129]. Another novel ATRi – ART0380 – is in a phase 1/2 clinical trial as a monotherapy or in combination with gemcitabine in patients with advanced or metastatic solid tumors (
Table 1).
ATR inhibition is effective in combination with other drugs that induce RS. For example, ATR inhibition by ceralasertib as a single agent and in combination with either CHK1 or WEE1 inhibitors was effective in several preclinical models of Mantle cell lymphoma and diffuse large B-cell lymphoma (DLBCL) regardless of their
TP53,
MYC, and
ATM mutational status in vitro and in vivo studies [
130]. Also high antitumor activity of ATRi (ceralasertib) and WEE1i (adavosertib) was shown in non-germinal center DLBCL cell lines, characterized by high
MYC expression and
CDKN2A/B deletion [
131]. ATR inhibition led to accumulation of 53BP1 nuclear bodies in daughter G1 cells and G1 arrest. WEE1 inhibition caused more pronounced DNA damage, inducing arrest in the S phase, and rapid induction of apoptosis.
In vivo xenograft DLBCL models showed potential for effective ATRi combinations. Moreover, ATRi and CHK1i are shown to resensitize PARPi-resistant,
BRCA1-deficient cancer cells to PARPi [
132,
133,
134,
135,
136] that also makes ATR-CHK1 pathway an attractive target in drug resistance context.
Particular challenge for anticancer therapy is to kill cells in a quiescent or slowly growing state. So it was demonstrated that treatment with the ATRi enhanced cell apoptotic signaling induced by cisplatin in quiescent cancer cells in vitro [
137].
Table 1.
Current clinical trials of ATRi.
Table 1.
Current clinical trials of ATRi.
Compound |
Study Phases |
Key Indications |
References |
Berzosertib (M6620, VX-970) |
Phase 2 (7 trials) |
Different types of cancer, including DDR deficient and TP53 mutant tumors |
NCT02595892 [100] NCT04266912 NCT03517969 NCT02567409 [138] NCT03896503 NCT04216316 NCT03641313 NCT03718091 (completed) NCT02487095 [139,140] |
Phase 1 (9 trials) |
Different types of cancer, including DDR solid tumors |
NCT02723864 NCT02589522 NCT02595931 NCT05246111 NCT04266912 NCT02567422 NCT02627443 NCT04216316 NCT04052555 NCT02157792 [94,95,96,101] |
|
|
|
Ceralasertib (AZD 6738) |
|
NSCLC |
NCT05450692 |
Phase 2 (28 trials) |
Different types of solid tumors, including NSCLC, breast and ovarian cancers |
NCT02264678 NCT04417062 NCT05061134 NCT05941897 NCT04564027 NCT05582538 NCT03801369 NCT04699838 NCT04090567 NCT03579316 NCT03878095 (suspended) NCT04239014 (withdrawn) NCT03334617 NCT03330847 NCT02937818 NCT03833440 NCT02813135 NCT04298021 NCT04298008 NCT03462342 NCT03428607 (completed) NCT03780608 NCT04065269 NCT04361825 NCT02576444 (terminated) NCT03740893 NCT03182634 NCT02664935 |
Phase 1 (13 trials) |
Different types of solid tumors and leukemias |
NCT05469919 NCT02264678 NCT05514132 NCT03328273 NCT03022409 (completed) NCT04704661 NCT03669601 NCT02630199 NCT03770429 NCT02223923 NCT01955668 (completed) NCT03527147 (completed) |
Elimusertib (BAY1895344) |
Phase 2 (1 trial) |
Relapsed or refractory solid tumors |
NCT05071209 |
Phase 1 (10 trials) |
Different types of carcinomas and lymphomas |
NCT05010096 (withdrawn) NCT03188965 (completed) NCT04095273 (completed) NCT05071209 NCT04616534 NCT04267939 NCT04491942 NCT04535401 NCT04576091 NCT04514497 |
Gartisertib (M4344, VX-803) |
Phase 2 (1 trial) |
Advanced breast cancer with DDR mutations |
NCT04655183 (withdrawn) |
Phase 1 (1 trial) |
Solid tumors |
NCT02278250 (completed) |
Camonsertib (RP-3500) |
Phase 1/2 (2 trials) |
Advanced solid tumors |
NCT04972110 NCT04497116 |
ART0380 |
Phase 2 (1 trial) |
Advanced tumors |
NCT05798611 |
Phase 1/2 (1 trial) |
Advanced tumors |
NCT04657068 |
CHK1 inhibitors (CHK1i) have actively been investigated in different tumor models and in combinations with a variety of drugs. The most promising compounds are prexasertib by Lilly Oncology, SCH 900776 by Merck and Co., and SRA737 by Sierra Oncology Inc. all of which are in early clinical trials (
Table 2).
Prexasertib (LY2606368, ACR-368) is a highly selective dual CHK1/CHK2 inhibitor that prevents CHK1 autophosphorylation, stabilizing CDC25A and increasing RS, leading to replication catastrophe and apoptosis [
141] (
Figure 3). It is effective in monotherapy and in combination with other replication-stress inducing agents such as PARPi, antimetabolites and platinum-based chemotherapy [
142]. The FDA has granted fast track designations to prexasertib in platinum-resistant ovarian cancer and endometrial cancer as monotherapy or in combination with low-dose gemcitabine (NCT05548296). As
MYCN amplification has been shown to increase RS it is considered as a possible additional biomarker for use of CHK1 inhibitors like prexasertib in neuroblastoma [
143]. Prexasertib was tested either as a single agent or in combination with PARPi olaparib in serous carcinoma PDX models and in a panel of ovarian cancer cell lines[
134,
144,
145]. Several phase 1 and 2 clinical trials are ongoing (
Table 2).
SRA 737 (PNT 737, CCT245737) is a novel orally bioavailable selective CHK1i that has shown preclinical activity in
MYC-amplified models of neuroblastoma [
146] and lymphoma [
147]. CHK1 inhibition by SRA 737 showed synthetical lethality with loss of B-family DNA polymerase function in lung and colorectal cancer cells [
148].
MK-8776 (SCH 900776) a highly selective dual CHK1/2i [
149]. This inhibitor was studied as a monotherapy and in combination with gemcitabine in patients with advanced solid tumors in phase 1 of clinical trials [
150]. MK-8776 is capable of restoring the sensitivity for chemotherapy drugs in cancer cells that overexpress P-glycoprotein, the ABC transporters which regulate the uptake and efflux of chemotherapeutics [
151].
Table 2.
Current clinical trials of CHK1i.
Table 2.
Current clinical trials of CHK1i.
Compound |
Study Phases |
Key Indications |
References |
Prexasertib (LY2606368, ACR-368) |
Phase 2 (7 trials) |
Different types of tumors, including small cell lung cancer, ovarian cancer, etc. |
NCT02735980 (completed) [152] NCT03414047 (completed) NCT02203513 (terminated) NCT02873975 (completed) NCT04095221 NCT04032080 (completed) NCT05548296 |
Phase 1 (14 trials) |
Different types of solid tumors and leukemias |
NCT02778126 (completed) NCT02514603 (completed) NCT03495323 (completed) NCT02860780 (completed) NCT01115790 (completed) NCT03057145 (completed) NCT04095221NCT02808650 (completed) NCT04023669 NCT05548296 NCT03735446 (terminated) NCT02649764 (completed) NCT02124148 (completed) NCT02555644 (completed) |
SRA 737 |
Phase 1/2 (2 trials) |
Advanced solid tumors or non-Hodgkin's lymphoma |
NCT02797964 [152,153] NCT02797977 |
MK-8776 (SCH 900776) |
Phase 2 (1 trial) |
Leukemias |
NCT00907517 (terminated) [154] |
Phase 1 (2 trials) |
Solid tumors, leukemias and lymphomas |
NCT00779584 (completed) |
The ATRi and especially CHK1i drugs have been extensively developed only in recent years, and available clinical data is relatively limited, compared to approved PARPi. Toxicity remains a major hurdle for ATRi and CHKi [
155]. More clinical data and basic research would hopefully allow to determine precise markers and indications for these drugs, allowing for the maximum therapeutic window.
2.2.2. PARP inhibitors
Presently many PARPi such as olaparib, rucaparib, niraparib and talazoparib are approved by the US FDA primarily for
BRCA1/2-mutated tumors [
172,
173]. Recent data about clinical trials of these drugs are represented in
Table 3. It is pivotal to verify
BRCA1/2 status of patients before therapy due to the role of HR in the potency of PARPi usage. However, even
BRCA1/2-mutated tumors can manifest resistance to PARPi. There are many ways to perform it such as replicative fork stabilization, HSP90-mediated BRCA1 stabilization and subsequent HR repair [
174], or recently researched recruitment of protein complex shieldin: REV7, RINN1, RINN2, and RINN3 (
Figure 3). This complex promotes NHEJ-dependent DNA repair by ATM-53BP1-RIF1-REV7 pathway that leads to the development of resistance to PARPi by the HR-independent pathway [
175,
176].
Some trials indicated that PARPi treatment may be improved by addition of ATRi [
177,
178]. That being the case, many up-to-date studies suggest that PARPi can be sufficiently effective against a wider variety of tumors than was thought before. Numerous clinical trials demonstrate beneficial results of PARPi treatment of pancreatic cancer [
179], urothelial carcinoma [
180], NCT03397394) and mesenchymal sarcomas [
181].
Previously, it was demonstrated that PARP inhibition therapy led to an increase of ATR and CHK1 phosphorylation, suggesting that activation of the ATR-CHK1 replication fork protection pathway is one of the main ways to save genome stability in response to PARP shortage. Hence, inhibition of ATR (ceralasertib) or CHK1 (MK-8776) in combination with olaparib led to a considerable synergistic effect that was proved by experiments on HGSOC both
in vitro and
in vivo [
132]. Interestingly, Parmar et al. [
134] demonstrated that combination therapy with olaparib and a CHK1i prexasertib was highly effective in models of ovarian and osteosarcoma cancer cells and patient-derived xenografts, resistant to PARPi monotherapy. The mechanism of synergy was associated with
RAD51 depletion and replicative fork destabilization; the best effect was achieved in combination treatment of
RAD51-mutated cells. Experiments on xenograft models (HGSOC) with
BRCA1 mutations and without them showed that PARPi monotherapy did not cause effect, but models were sensitive to CHK1i prexasertib monotherapy. Combination of PARPi and CHK1i showed increased efficiency compared to CHK1i monotherapy.
Another growing field for PARP inhibitors usage is combination treatment with WEE1 inhibitors such as a novel small molecule adavosertib (AZD1775). It might be connected with the G2/M checkpoint, which is vital for correct DNA repair before mitosis and further cell cycle progress. Inhibition of WEE1 compromises G2 arrest, leading to abnormal exit to mitosis, and accumulation of DNA damage, leading to RS and subsequent cell death. Thus , it was recently demonstrated [
182] that WEE1i adavosertib monotherapy of TNBC was effective, inhibiting RAD51-mediated HR DNA repair, and increasing quantity of γH2AX foci.cExperiments on breast cancer cell lines (MDA-MB-231, BT-549) showed synergistic effects with combination of PARPi olaparib and adavosertib. During combinational treatment, the number of DNA-damaged cells was greater than in adavosertib monotherapy; such synergy was caused by HR deficiency that noticeably enhanced PARPi impact. Besides, the same study demonstrated highly efficient synergism of tumor growth inhibition by PARPi and WEE1i combined treatment in human breast cancer xenograft models (MDA-MB-231) without significant toxicity. Similar results were also reported by [
183], emphasizing G2-M arrest induced after PARP inhibition by talazoparib. PARP inhibition led to increased expression or phosphorylation of major proteins involved in S and G2 DNA damage checkpoints: Cyclin B1, Rb, WEE1, CDK1, FOXM1, CHK1, CHK2 and ATM. Interestingly, sequential therapy did not reduce the efficiency of PARPi and WEE1i combination for ovarian cancer cells in vitro and in vivo, compared to concurrent inhibition, while reducing toxicity for non-transformed cells.
Table 3.
Clinical trials of approved PARPi.
Table 3.
Clinical trials of approved PARPi.
Compound |
Study Phases |
Key Indications |
References |
Olaparib |
Phase 4 (4 trials) |
Ovarian cancer (3 trials), prostate cancer (1 trial), metastatic breast cancer (1 trial) |
[156,157,158,159] |
Phase 3 (37 trials) |
Ovarian cancer (more than 30 trials), breast cancer (13 trials), prostate cancer (4 trials) |
Phase 2 (more than 200 trials) |
Ovarian cancer (more than 30 trials), breast cancer (more than 30 trials), prostate cancer (more than 20 trials), lung cancer (more than 20 trials) |
Phase 1 (more than 100 trials) |
Ovarian cancer (more than 30 trials), breast cancer (more than 20 trials), prostate cancer (12 trials), lung cancer (10 trials) |
Niraparib |
Phase 4 (3 trials) |
Ovarian cancer (3 trials) |
[160,161,162,163] |
Phase 3 (23 trials) |
Ovarian cancer (12 trials), fallopian tube cancer (5 trials), prostate cancer (3 trials), breast cancer (2 trials), |
Phase 2 (more than 100 trials) |
Ovarian cancer (more than 30 trials), breast cancer (15 trials), fallopian tube cancer (9 trials) |
Phase 1 (62 trials) |
Ovarian cancer (19 trials), breast cancer (13 trials), prostate cancer (7 trials) |
Rucaparib |
Phase 3 (8 trials) |
Ovarian cancer (4 trials), fallopian tube cancer (4 trials), prostate cancer 2 trials) |
[164,165,166,167] |
Phase 2 (40 trials) |
Ovarian cancer (8 trials), prostate cancer (7 trials), breast cancer (4 trials), |
Phase 1 (24 trials) |
Ovarian cancer (7 trials), breast cancer (4 trials), prostate cancer (4 trials) |
Talazoparib |
Phase 3 (5 trials) |
Ovarian cancer (2 trials), breast cancer (1 trial), prostate cancer (1 trial) |
[168,169,170,171] |
Phase 2 (64 trials) |
Breast cancer (17 trials), prostate cancer (8 trials), ovarian cancer (4 trials) |
Phase 1 (51 trials) |
Breast cancer (14 trials), prostate cancer (5 trials), ovarian cancer (4 trials) |
2.2.3. WEE1 and PKMYT1 inhibitors
WEE1 and PKMYT1 (MYT1) are two protein kinases that regulate activity of CDK complexes through inhibitory phosphorylations. WEE1 inhibits activity of CDK2 at the both G1/S and G2/M transitions, while PKMYT1 is active only in the G2/M checkpoint. Both are rarely mutated in cancers and in tumors with high levels of RS they act as oncogenes, protecting cells from excessive DNA damage. Both are overexpressed in many hematological and solid tumors [
209].
Currently, the most commonly investigated WEE1i is a small molecule AZD1775 (adavosertib,
Table 4). Presently it is in trials as an anticancer drug for different types of solid tumors even in pediatric patients [
199]. The most recent studies involved in phase I or phase 2 of clinical trials are examining WEE1i for the treatment of pancreatic, gastric, head and neck, breast, ovarian, and other tumors [
194]. Many therapies with WEE1i frequently include combinations with other drugs that induce RS, such as carboplatin [
184], gemcitabine [
186], and PARPi like olaparib [
210].
WEE1 inhibition by adavosertib in tumor cells causes acceleration of cell cycle promotion by activation of CDK2 that, in turn, leads to RS, DNA aberrations and further cell death. Other research by Lindemann and colleagues [
211] suggested that DNA aberrations and RS, caused by WEE1 inhibition, could be useful for cancer treatment under conditions of DNA repair disorder. Сombined incubation with Rad51 inhibitor B02 and adavosertib manifested notable synergism, with increased markers of DNA damage (γH2AX) and RS (pRPA32), and levels of cell death, than in monotherapy. As was demonstrated in previous works [
212], CHK1 phosphorylation level was decreasing, while CDK1 activity was rising that, as a result, caused accumulation of RS, indicating DSB repair shortage. Interestingly, in HPV-positive lines, an increase of p53 level was observed that can be connected with activity of E6 or E7 oncogenes [
213], although p21 level was increased irrespective of HPV status in response to combination of B02 and adavosertib.
In vivo experiments in mouse models of oral tongue cancer have shown that HPV-negative tumors were not sensitive to B02 and adavosertib mono- or combined therapy. In HPV-positive mice, drugs’ combination significantly inhibited growth of the tumor and substantially increased animal survival rate [
211]. These findings are very important for the therapeutic aims due to the role of Rad51 in patients’ survival rate [
214].
P53 status is found to play a significant role in WEE1i performance. Thus,
TP53-depleted HNSCC cells demonstrate remarkable accumulation of SSB and DSB DNA damage markers and PARP1 cleavage in response to adavosertib, compared to
TP53 WT (wild type) cells. Interestingly, the number of 53BP1 foci, which is an important marker of DNA damage, was lower in
TP53 knockdown cells, however most of the 53BP1-positive cells did not express γH2AX. It can be considered as a mismatch between 53BP1 foci localization and DSBs [
215]. Furthermore, previous study investigated that WEE1i sensitized
TP53-mutated mouse xenografts to cisplatin exposure, indicating potency of adavosertib as an effective supplemental drug for
TP53-mutated tumors therapy [
216]. Data from the previously mentioned article [
131] revealed that adavosertib and ATRi ceralasertib treatments slowed progression of the replication fork and increased origin firing. WEE1i led to activation of the ATR–CHK1 pathway and decrease of CHK1 and ATM protein expression after 24 hours. However, the combination of adavosertib and ceralasertib was not effective
in vivo, resulting in tumor regressions comparable to the adavosertib single-agent.
PKMYT1 is overexpressed in a number of tumors with markers of RS and PKMYT1 inhibitors (PKMYT1i) are effective in preclinical
in vitro and
in vivo models. PKMYT1 inhibition was effective in
MYCN-amplified neuroblastomas, but not neuroblastomas without amplification [
217]. Blocking PKMYT1 activity was effective in eradication of
CCNE1-amplified ovarian cancer cells, but not cell lines without amplification through preventing completion of DNA synthesis and increasing the rates of premature mitotic entry [
38]. Notably
PKMYT1 is overexpressed in
CCNE-amplified ovarian carcinoma [
218], and currently PKMYT1i RP-6306 is in clinical trials in this setting (
Table 4).
Additionally, PKMYT1 and WEE1 inhibition synthetically eradicates cancers with high levels of RS such as glioma [
219] and HGSOC, relatively sparing normal tissues or cancers with lower levels of RS [
220]. As WEE1i are hindered by toxicity, the authors consider such combinations more selective for therapy. As PKMYT1, but not WEE1, is more important for G2/M transition during checkpoint recovery [
221] its inhibitors could be important for a number of combinational therapies aimed at RS.
Table 4.
Clinical trials of WEE1 and PKMYT1 inhibitor.
Table 4.
Clinical trials of WEE1 and PKMYT1 inhibitor.
Compound |
Study Phases |
Key Indications |
References |
Adavosertib (AZD1775) |
Phase 2 (32 trials, including 7 terminated and 1 withdrawn) |
Solid tumors, harboring CCNE1 amplification, ovarian (2 studies), neuroblastoma, medulloblastoma, and rhabdomyosarcoma |
[184,185,186,187,188,189,190,191,192,193] |
Phase 1 (34 trials, including 6 terminated and 1 withdrawn) |
HNSCC, uterine cancers, TNBC, pancreatic cancer, acute myeloid leukemia, glioblastoma |
[194,195,196,197,198,199,200,201,202,203,204,205,206,207,208] |
2.3.4. CDK inhibitors and RS
Сyclin-dependent kinases in complexes with their Cyclins regulate several critical processes in the cell. The better-known group, consisting of CDK1-6 primarily controls the transition through stages of the cell cycle, while other CDKs, such as CDK7, CDK8/19, CDK9 and CDK12/13 are mainly involved in transcription [
222]. Interestingly both groups of CDKs are implicated in RS and DNA damage response, as proper alignment of transcription and cell cycle transition is critically important to the proper replication process (
Figure 3). As inhibitors of CDKs are approved by the regulator or are investigated in clinical trials, more studies focus on their impact on tumors with increased RS (
Table 5).
CDK1 is the Cyclin-dependent kinase essential for cell cycle progression in S, G2 and M phases of the cell cycle and can also replace other Cyclin-dependent kinases in many models [
13,
223]. CDK1 inhibition compromises the BRCA1-dependent ATR and ATM DDR in the S phase, increasing sensitivity to DNA damaging agents [
224]. Compromised CDK1 activity also leads to increased sensitivity to PARPi [
225]. On the other hand CDK1 inhibition is an important mechanism, limiting transcription-replication conflicts [
11].
As discussed above, CDK2/Cyclin E is responsible for G1/S entry by phosphorylating Rb and components of the replication machinery such as CDC6, treslin and RECQL4, as well as CDT1 and MCM complex components [
30]. Amplification of the
CCNE1, is common in many cancers, especially in breast and ovarian carcinomas. Increased activity of CDK2/Cyclin E, in these tumors is known to increase RS and genetic instability via several mechanisms such as shortening of the G1 phase and aberrant origin licensing [
30]. Although
CCNE-amplified tumors are more prone to RS they remain hard to treat using standard DNA-damaging chemotherapy regimens, especially in ovarian cancer. Several studies revealed, counterintuitively, that CDK2 participates in DNA repair [
5,
226], and
CCNE-amplified ovarian carcinomas rely on CDK2 for DNA repair through homologous recombination, its inhibition compromises replication fork repair [
5]. The same study also demonstrated that Cyclin E1 is present at the stalled forks, probably participating in repair.
CDK4/6 inhibitors (palbociclib, ribociclib, abemaciclib) are known as effective drugs for breast cancer treatment irrespective of the p53 status [
227,
228]. Long-term G1 arrest induced by palbociclib caused RS that was a reason for significant cell proliferation decrease due to transition to senescent state of
TP53 WT cells or, in the absence of p53, by causing cells to undergo mitotic catastrophe, resulting in DNA damage [
229]. This difference in cells' fate depended on the level of p21 – one of the main proteins involved in transition to a senescence condition. Thus, in WT cells, p53-induced p21 level rise, while in
TP53 KO (knockout) cells induction was absent. Effect of palbociclib for
TP53 WT and especially
TP53 KO was ATR-dependent: ATR inhibition after CDK4/6i 7 days treatment demonstrated an increase in the number of fragmented nuclei that is a consequence of chromosome segregation errors. These data suggested that prolonged CDK4/6 inhibition led to a shortened period of replication and increased the number of cells, which enter mitosis.
Transcriptional CDKs are also implied in RS regulation. CDK8 and CDK19 are two homologous kinases which regulate initiation of transcription as part of the Mediator complex. They are required for transcription activated by a number of transcription factors, such as STATs, SMADs, beta-catenin, p53 and others [
230]. Similarly to CDK2, CDK8/19 have a dual role in response to RS. One article demonstrates that CDK8/19 deletion decreases RS due to replication-transcription conflicts [
231], and CDK8/19 activity is required for sensitivity to ATRi and CHK1i. On the other hand, in uterine fibroids inhibition of CDK8/19 increased the number of stalled replication forks and markers of RS, including ATR phosphorylation. This phenotype was also dependent on R-loop formation [
232]. Additionally, at least, in certain cancers such as prostate carcinoma inhibition of CDK8/19 led to increased ATR-dependent RS and DNA damage by inducing aberrant G1/S transition [
233]. Another recent study has demonstrated a similar increased G1/S transition in CML [
234]. CDK8/19 is also required for normal origin firing during replication by interacting with the MTBP (Mdm2 p53 binding protein) complex with Treslin, and its inhibition leads to an increase in the number of fragile metaphase chromosome sites [
235]. It is possible that similarly to CDK2, CDK8/19 participates in ATR-alternative repair pathways, during RS, as DNA-repair proteins such as BRCA2 and MDC1 were identified as CDK8/19 substrates [
236], and inhibition of CDK8/19 increased activity of DNA-damaging agents [
237].
While CDK8/19 are involved in initiation of transcription, CDK12 and CDK13 regulate transcription termination and splicing. CDK12/13 are involved in transcriptional regulation of a number of DNA damage response genes, such as
BRCA1,
ATR and
FANC1 [
238]. CDK12 expression is required for expression of core replication genes, and its inhibition delays G1/S transition and increases in the number of chromosomal aberrations. This replication-dependent DNA damage is caused by reduced processivity of RNA polymerase II in long poly-(A)-signal-rich genes [
239]. CDK12 was implicated in sensitivity to PARPi [
240] as well as survival of ovarian cancer cell lines irrespective of synergy with PARPi [
241].
Table 5.
Clinical trials of CDK2 inhibitors in tumors with RS markers.
Table 5.
Clinical trials of CDK2 inhibitors in tumors with RS markers.
Compound |
Study Phases |
Key Indications |
References |
BLU-222 |
Phase 1/2 (1 trial) |
Solid tumors, including CCNE1-amplified, ovarian carcinoma, breast cancer, endometrial and gastric cancer |
NCT05252416 |
INX-315 |
Phase 1/2 (1 trial) |
Solid tumors, including breast cancer who progressed on a prior CDK4/6i regimen, and CCNE1-amplified solid tumors |
NCT05735080 |
PF-07104091 |
Phase 1/2 (2 trials) |
Small cell lung cancer, ovarian cancer, breast cancer |
NCT04553133 NCT05262400 |
ARTS-021 |
Phase 1/2 (1 trial) |
CCNE1-amplified solid tumors |
NCT05867251 |
INCB123667 |
Phase 1 (1 trial) |
Solid tumors |
NCT05238922 |