Abstract
Purpose:
CDK12 is a cyclin-dependent kinase (CDK) that is mutated or amplified in multiple cancers. We previously described a subtype of prostate cancer (PC) characterized predominantly by frameshift, loss-of-function mutations in CDK12. This subtype exhibits aggressive clinical features.
Experimental Design:
Using isogenic PC models generated by CRISPR/Cas9-mediated inactivation of CDK12, we conducted a chemical library screen of ~1800 FDA-approved drugs. We inhibited cyclin K and CDK13 and evaluated the effects on poly ADP-ribose polymerase inhibitor (PARPi) sensitivity. CDK12 truncation and kinase domain mutations were expressed in cell lines to determine effects on PARPi sensitivity. Mice bearing control and CDK12 mutant prostate tumors were treated with rucaparib. Finally, we evaluated prostate specific antigen (PSA) responses in patients with CDK12 mutations treated with rucaparib on the TRITON2 trial.
Results:
Cancer cells lacking CDK12 are more sensitive to PARPi than isogenic wild-type cells, and sensitivity depends on the degree of CDK12 inhibition. Inhibiting cyclin K, but not CDK13, also led to PARPi sensitivity and suppressed homologous recombination. CDK12 truncation mutants remained sensitive to PARPi, whereas kinase domain mutants exhibited intermediate sensitivity. The PARPi rucaparib suppressed tumor growth in mice bearing CDK12-mutated tumors. Finally, 6 of 11 (55%) PC patients with biallelic CDK12 mutations had reductions in serum PSA levels when treated with rucaparib on the TRITON2 clinical trial.
Conclusions:
In PC, sensitivity to PARPi is dependent on the specific type and zygosity of the CDK12 mutation. PARPi monotherapy may have some activity in PC patients with biallelic inactivating CDK12 alterations.
INTRODUCTION
While drugs targeting a subset of driver oncogenes, such as EGFR or NTRK fusions, have shown tremendous clinical benefit (1,2), most oncogenes and tumor suppressors are not yet targetable. In patients whose tumors harbor loss of function mutations in the canonical homologous recombination (HR) repair genes, BRCA1 and BRCA2, poly ADP-ribose polymerase (PARP) inhibitors (PARPi) have shown benefit (3). This synthetic lethality is observed across multiple tumor types, as recent clinical trials in ovarian, breast, pancreatic and prostate cancers have all shown clinical efficacy (4–7). However, whether alterations in genes other than BRCA1/2 also confer PARPi sensitivity remains an open area of investigation (8).
Although the androgen axis plays a key role in the pathogenesis and treatment of prostate cancer (PC), multiple genomics studies have found that alterations in other genes and pathways are common in advanced PC (9–11). We and others previously identified that loss of function, frameshift and point mutations in cyclin dependent kinase-12 (CDK12) are found in approximately 7% of advanced PC patients. Mutations in CDK12 are associated with aggressive clinical features, including a higher Gleason score at diagnosis and shorter time to developing castration-resistant prostate cancer (CRPC) and metastatic disease (12,13). CDK12 is a transcription-associated CDK and regulates the elongation phase of transcription by phosphorylating the carboxy-terminal domain (CTD) of RNA polymerase II (14,15). Moreover, CDK12 regulates gene expression through controlling alternative splicing (16) and suppressing intronic polyadenylation (17). Interestingly, these effects on gene expression appear to preferentially affect long genes (17,18). We and others previously showed that genetic or chemical inhibition of CDK12 leads to loss of DNA damage repair genes, including BRCA1, which leads to a functional HR deficiency (18–20). However, although olaparib and talazoparib have been recently approved for CDK12-mutated prostate cancer, controversy remains as to the magnitude of the effect and whether all patients with CDK12 alterations are equally sensitive to PARPi (21,22). Given that models of CDK12-deficient prostate cancer are lacking, generating pre-clinical models of this molecular PC subtype and identifying therapeutic strategies for CDK12-deficient cancers are critical.
Here, we generated CDK12 knockdown (KD) and knockout (KO) isogenic cancer cell lines using CRISPR/Cas9, performed a small molecule screen of over 1,800 bioactive and FDA-approved compounds and demonstrated that CDK12 deficient cells are sensitive to multiple DNA damage agents. Focusing on PARPi, we showed that these drugs selectively kill CDK12 KO cells in vitro and in vivo, and that loss of CDK12 or its binding partner, cyclin K (CCNK), but not CDK13, led to PARPi sensitivity. Finally, we evaluated PSA responses to rucaparib monotherapy in advanced, heavily pre-treated, CRPC patients with tumors harboring multiple types of CDK12 alterations.
MATERIALS AND METHODS
Animal studies.
NSG (NOD/SCID/gamma) mice were housed in the UCSF barrier facility under pathogen-free conditions and were obtained through an in-house breeding core. For tumor cell line xenografts, 1× 106 cells were injected subcutaneously into each flank (bilateral) of 7–10 week old male NSG mice. The injected cells were resuspended in 1:1 serum-free media and Matrigel (BD Biosciences). Tumor measurements were performed once per week with digital calipers, and the volume of each tumor was calculated based on the formula: V=0.52*length*width2, where the width represents the shorter axis. Mice were enrolled into treatment groups once tumor volumes reached between 50–150 mm3. Rucaparib camsylate (Clovis) was reconstituted in 0.5% methylcellulose (Spectrum) as per the manufacturer’s recommended concentration and administered by oral gavage (per os) at 150 mg/kg daily (Monday – Friday) for 3 weeks.
Cell culture.
LnCAP, PC3, C42B, TRAMP-C2 and DLD-1 cells were obtained from the ATCC. OVCAR8, CAOV3 and MDA-MB-231 cells were obtained from the UCSF Cell Culture Facility. Cells were grown in standard conditions with antibiotics (RPMI-1640 with 10% FBS for LnCAP, PC3, C42B, DLD-1, OVCAR8 and CAOV3 cells; DMEM with 10% FBS for MDA-MB-231 and TRAMP-C2 cells) and routinely tested for mycoplasma every 3 months (Lonza MycoAlert Mycoplasma Detection Kit) throughout the study, and most cell lines were lasted tested in November 2023. Cell lines were all validated by STR DNA profiling (UC Berkeley DNA Sequencing Facility), and were used within 40 passages from the time of STR DNA profiling for all experiments shown.
Plasmids and cloning.
pSpCas9(BB)-2A-GFP (PX458) (Addgene plasmid # 48138) and lentiGuide-Puro (Addgene plasmid # 52963) were gifts from Feng Zhang. The pHR-SFFV-dCas9-KRAB-BFP and pCRISPRia_v2 plasmids (Addgene plasmid #46911 and 84832) were gifts from Drs. Stanley Qi, Luke Gilbert and Jonathan Weissman. The pDONR221 plasmid was from Thermo Fisher Scientific. Cloning was performed using standard protocols. sgRNA sequences to target CDK12, CDK13, and CCNK are provided in Supplementary Table S3. Gel purification was performed using the NucleoSpin Gel and PCR Clean-up kit (Takara Bio Inc.) according to the manufacturer’s protocol. Annealed sgRNAs were ligated into the plasmid using the Quick Ligation Kit (New England BioLabs) according to the manufacturer’s protocol. For Gateway cloning, BP and LR Clonase II (Thermo Fisher Scientific) reactions were performed according to the manufacturer’s protocol.
Generation of CRISPR Interference (CRISPRi) knockdown cells.
CRISPRi cell lines were generated as previously described2. Briefly, cells were transduced with dCas9-KRAB-BFP and sorted by FACS (BD Fusion) for BFP positivity. Cells were expanded and then sorted at least once again for BFP to obtain an optimized pure population. The dCas9-KRAB expressing cells were then transduced with lentivirus containing the sgRNA targeting the gene of interest. After 2–3 days, cells were selected in 3–5 μg/ml of puromycin for at least 3 days. Cells were used for experiments up to 8 passages and kept in puromycin to maintain a stable knockdown of the gene of interest.
Generation of CDK12 and CDK13 knockout cells.
Cells were transfected with the PX458 plasmid containing the guide targeting the gene of interest using Lipofectamine 3000 (Thermo Fisher) according to manufacturer’s instructions. 48 – 72 hours after transfection, cells were sorted for green fluorescence protein (GFP) expression, and then into single cells in a 96-well plate format. Clones were expanded and validated by Western blotting and sequencing the target site.
Small molecule compound screening.
50 uL cell suspension/250 cells per well were seeded into 384 well white plates (Greiner) using a WellMate bulk dispenser and incubated overnight prior to addition of 50 nL Bioactive Small Molecule Library (SelleckChem) by 384 pin tool (VP Scientific). Stock compound concentrations were 10, 1 and 0.1 mM in DMSO, yielding final assay concentrations of 10, 1 and 0.1 μM (0.1% DMSO). Compounds were pre-arrayed in columns 3 through 22 (320 per plate), with columns 1, 2, 23 and 24 (untreated negative controls, n=32) receiving DMSO only. Matched plate pairs (control reference and CDK12 KO) were assayed for viability after 72 and 144 hours using Promega CellTiter-Glo according to the manufacturer’s instructions. Luminescence was recorded using an Envision Xcite plate reader (Perkin Elmer).
Data normalization.
Compound luminescence counts were normalized to fold change values using respective plate mean negative controls (n = 32). Per plate significance limits were obtained from mean negative control 3 x SD of the mean, with compound fold change values < Mean-3SD curated as significantly outside control variation (viability hits).
Data reduction.
Fold change ratios (Control/CDK12 KO) were calculated to identify compounds with selectivity relative to reference genetic background. Ratio significance limits were established using aggregate mean +/− 3 x SD of the mean for all normalized results, with ratios > Mean Ratio+/− 3SD indicating significantly higher selectivity in CDK12 KO cells.
Drug dose-response assays.
Cells were seeded in a 96-well plate at a density of 1,000 cells per well (C42B, PC3, DLD-1, MDA-MB-231) or 2,000 cells per well (LNCaP, CAOV3, OVCAR-8). After 24 hours, drug or DMSO was added to the cells. The following drugs and concentrations were used: rucaparib, olaparib and veliparib (4-fold serial dilutions, starting at 50μM to 0.763nM), and talazoparib (4-fold serial dilutions starting from 10μM to 0.153nM). Cells were exposed to drug for seven days. On the last day of drug treatment, cells were incubated with Cell Proliferation Reagent WST-1 solution (Roche) according to the manufacturer’s protocol. Absorbance values were obtained using an Epoch Microplate Spectrophotometer with Gen5 software, normalized to the DMSO control, and values were plotted and analyzed using Prism 9 software (GraphPad).
Lentiviral production.
Lentiviral production was carried out using calcium-phosphate-mediated transfection of HEK293T-Lenti-X cells using standard procedures. Lentivirus was concentrated using the Lenti-X concentrator (Takara Bio Inc.) according to the manufacturer’s protocol. Stably transduced cells were selected with puromycin or blasticidin for at least 5 days or selected by FACS.
Flow cytometry.
Cells were trypsinized using standard procedures. Prior to sorting, cells were passed through a 40μm cell strainer. For single cell cloning, single cells were sorted into individual wells in a 96-well plate and expanded over 3–6 weeks. Analysis and cell sorting were performed on the Attune analyzer or FACS Aria III (Becton Dickinson), and analysed using FlowJo (TreeStar) or FACSDiva software (BD Biosciences).
Quantitative PCR (QPCR).
Total RNA was isolated from cells using the Quick RNA Kit (Zymo Research). cDNA was synthesized using the Superscript III RT First Strand Kit (Invitrogen). QPCR was performed using SYBR Green master mix (Roche) in an Applied Biosystems QuantStudio7 machine. Ct values were normalized to actin and GAPDH, and relative expression was calculated using the 2ddCt method. Primer sequences for QPCR were found using the Harvard Primer Bank and are detailed in Supplementary Table S3.
Immunostaining and histology.
Tissue samples were fixed in 4% PFA overnight and paraffin embedded. Standard hematoxylin and eosin (H&E) staining was performed for routine histology. Antigen retrieval for immunohistochemistry was performed using the BioSB TintoRetriever and citrate buffer. The TSA Amplification Kit (Perkin Elmer #NEL700001KT) was used according to manufacturer’s specifications. Primary antibodies were incubated overnight and secondary antibodies were incubated for 2 hours. The following antibodies were used at the indicated concentrations: CDK12 (Atlas #HPA008038, 1:100), Phospho-Histone H3 (Cell Signaling #9701, 1:200), AR PG-21 (Millipore #06–680, 1:500), γH2AX (Cell Signaling #9718, 1:500), Normal Rabbit IgG (#3900, 1:100), Biotinylated anti-rabbit (Jackson #111-065-144, 1:1000). The DAB developing kit (Vector Laboratories) was used according to instructions.
Western blotting.
Cells were lysed in RIPA buffer the Halt protease and phosphatase inhibitor cocktail (Thermo Fisher Scientific). Protein concentration was measured using the BCA Protein Assay Kit (Thermo Scientific). Lysates were mixed at a 1:1 ratio with Laemmli Buffer (Bio-Rad) supplemented with beta-mercaptoethanol, boiled at 95°C, and then subjected to SDS-PAGE, transferred to PVDF membranes, blocked in 5% w/v BSA, incubated with primary antibody overnight. The next day, Horseradish Peroxidase (HRP) conjugated secondary antibodies were used and blot was visualized using ECL Detection Reagents (Genesee Scientific). Antibodies used include: CDK12 (Atlas #HPA008038, 1:1000 and Cell Signaling #11973, 1:1000), RNA Pol II pSer2 (Active Motif, 1:5000), Lamin B1 (Cell Signaling, 1:1000), GAPDH (Cell Signaling, 1:2000), CDK13 (Bethyl Laboratories, 1:1000), and cyclin K (Bethyl Laboratories, 1:1000). Uncropped blots with ladder markers are provided as Supplementary Fig. S6.
Neutral comet assay.
Neutral comet assay was performed according to the Trevigen protocol. Briefly, cells were trypsinized, combined with low melting agarose and spread on comet slides. Cells were lysed in Lysis Solution for 1 hour at 4°C in the dark, then immersed in neutral electrophoresis buffer for 30 minutes. Comets were ran in a large electrophoresis tank at 21 volts for 45 minutes at 4°C in the dark, then precipitated for 30 minutes at room temperature. Samples were washed in 70% ethanol and dried until flat, then stained with SYBR Gold. Slides were washed, dried and mounted in Prolong Gold, and imaged on a fluorescence microscope. 100–150 comets were imaged per sample, and Tail Moment was calculated using OpenComet in ImageJ.
Clinical trial data (TRITON2).
TRITON2 is an open-label, phase II study evaluating the safety and efficacy of rucaparib in men with mCRPC associated with DDR deficiency. This trial was conducted at 144 centers in 12 countries. Eligible patients at least 18 years old had histologically or cytologically confirmed adenocarcinoma or poorly differentiated carcinoma of the prostate that was metastatic and that had progressed on second-generation, AR-directed therapy (e.g., abiraterone acetate, enzalutamide, or apalutamide) for prostate cancer and one prior line of taxane-based chemotherapy for mCRPC. Disease progression on prior therapy was based on any of the following criteria: rise in PSA (minimum of two consecutive rising levels, with an interval of 1 week or more between each determination; the most recent screening measurement must have been greater than or equal to 2 ng/mL); transaxial imaging with new or progressive soft tissue masses on CT or MRI as defined by RECIST version 1.1; or radionuclide bone scan with at least two new metastatic lesions. Patients were surgically or medically castrated, with serum testosterone levels of 50 ng/dL or less. Patients with and without measurable visceral or nodal disease per RECIST criteria were eligible for the study; patients without measurable disease were required to have PSA levels of greater than 2 ng/mL on the most recent measurement. Patients had an Eastern Cooperative Oncology Group Performance Status of 0 or 1 and adequate organ function. Patients who had prior treatment with any PARP inhibitor, mitoxantrone, cyclophosphamide, or any platinum-based chemotherapy were excluded.
Patients were screened for the presence of a deleterious somatic or germline alteration in BRCA1, BRCA2, ATM, BARD1, BRIP1, CDK12, CHEK2, FANCA, NBN, PALB2, RAD51, RAD51B, RAD51C, RAD51D, or RAD54L through central genomic testing of plasma or tumor tissue (archival or contemporaneous), or through local testing. Central testing was performed by Foundation Medicine, Inc., and mutations in CDK12 were determined by monoallelic, biallelic or gene rearrangements.
Patients received oral rucaparib 600 mg twice daily until confirmed radiographic disease progression as assessed by investigator on the basis of modified RECIST and/or Prostate Cancer Clinical Trials Working Group 3 (PCWG3) criteria, unequivocal clinical disease progression, death, or other reason for discontinuation. Dose reductions were permitted if a patient had a grade 3 or greater or a persistent grade 2 adverse event (AE).
Study approval.
All animal experiments were performed at UCSF, and reviewed and approved by the UCSF IACUC. The TRITON2 clinical trial was approved by national or local institutional review boards and was carried out in accordance with the Declaration of Helsinki and Good Clinical Practice Guidelines of the International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use. Patients provided written informed consent before participation. In terms of the Representativeness of Study Participants, the demographics of the study population, which included men with prostate cancer, were previously reported (23,24).
Statistical analysis.
Statistical analysis was performed using Prism 9 software (GraphPad Software). All data are presented as mean +/− SEM, unless otherwise stated. When two groups were compared, the two-tailed Student t -test was used, unless otherwise stated. When three or more groups were compared, the one-way analysis of variance (ANOVA) test was used, followed by multiple testing with correction (Sidak) to determine significance between groups, unless otherwise stated. We considered p <0.05 as significant.
Data availability statement.
The whole exome sequencing (WES) data generated in this study are publicly available in the NIH Sequence Read Archive (SRA) under BioProject ID numbers PRJNA1145981 and PRJNA932332. Data from the TRITON2 clinical trial are available upon request from the corresponding authors. Drug screening data and western blots can be found within the article and its supplementary data files, and raw data for plots are available upon request from the corresponding authors. Plasmids and cell lines described in this study are available upon request from the corresponding authors.
RESULTS
A small molecule screen in CDK12 KO prostate cancer cells identifies multiple PARP inhibitors amongst the top selective hits
To generate pre-clinical prostate cancer models of CDK12-deficiency, we first generated single-cell derived CDK12 knockout (KO) clones using CRISPR/Cas9 targeting exon 1 of CDK12 in multiple prostate cancer cell lines, including LNCaP and C42B cells. These clones were confirmed to be deficient in CDK12 by western blotting (Fig. 1A–1B). These cells were further validated by whole-exome sequencing (WES) to demonstrate that we had successfully generated indels in exon 1 of CDK12 in LNCaP and C42B cells, leading to a frameshift and early truncation, which recapitulate the frameshift mutations found in CRPC patients (Supplementary Fig. S1A–S1C) (12). In cell culture, the CDK12 KO cells grew slightly slower than the isogenic wildtype control cells (Supplementary Fig S2A–S2B).
Figure 1: CDK12 deletion in prostate cancer cells is synthetic lethal with PARP inhibition.
(A-B) Western blots (WB) showing CDK12 protein in LNCaP (A) and C42B (B) CDK12 KO clones. Lamin B1 is shown as loading control. (C) Small molecule screen showing the fold change (FC) per drug in the C42B CDK12 KO (y-axis) over the FC in the Control (x-axis). The concentration of each drug is shown in cyan (0.1 μM), green (1 μM) and orange (10 μM), and the relative FC ratio is indicated by the size of the circle. The drug hits are indicated in the dotted square in the bottom right corner. (D) The drug hits are organized by drug class categories and are highly enriched in molecules involved in DNA damage. (E-F) Dose response assays to olaparib in LNCaP (E) and C42B cells (F). A representative assay of n=3 independent experiments performed in technical triplicates is shown for each cell line. (G-H) Clonogenic assay and quantification in C42B and CDK12 KO cells treated with the indicated doses of olaparib. The assay was performed in triplicate, n=3 independent experiments, *** indicates p<0.001, **** indicates p<0.0001. Error bars represent SEM for E-F. ANOVA with multiple comparison correction (Sidak) was performed for H.
To identify compounds selectively targeting CDK12-deficient cells, we utilized an isogenic pair of C42B cells to screen a commercially-available library containing approximately 1800 bioactive, FDA-approved compounds at 3 concentrations (10 μM, 1 μM and 0.1 μM). We used CellTiter-Glo and calculated the ratio of the control cell luminescence to CDK12 KO cell luminescence (after subtracting the background) for each compound and concentration and identified candidates that selectively killed the CDK12 KO if the ratio was 2. We focused on the 168 hour time point which had more hits. We found that among the top hits, multiple compounds belonged to the DNA damage subclass (Supplementary Table 1, Fig. 1C–1D). We were particularly intrigued that among the DNA damage compounds, 2 PARP inhibitors (PARPi), olaparib and rucaparib, were in the top 20 hits; a third PARPi (AG14361), which is chemically related to rucaparib, was also a significant top 40 hit. Given the recent approval of PARPi for CRPC and several ongoing clinical trials investigating the use of PARPi for CDK12-mutated cancers (NCT03012321, NCT04030559 and reviewed in (25)), we validated that CDK12 KO cells were indeed more sensitive to olaparib using dose-response and clonogenic assays (Fig. 1E–1H). We also evaluated the sensitivity to other PARPi, including talazoparib, rucaparib and veliparib, and found that CDK12 KO cells were more sensitive to these other PARPi as well (Supplementary Fig. S2C–S2E), which preferentially inhibited growth in the CDK12 KO cells (Supplementary Fig. S2F). Mouse TRAMP-C2 prostate cancer cells deficient in Cdk12 were also more sensitive to PARPi (Supplementary Fig. S2G–S2H). Taken together, these results demonstrate that CDK12 KO human and mouse prostate cancer cells exhibit enhanced sensitivity to multiple PARP inhibitors.
CDK12 loss in multiple cancer cell types results in PARPi sensitivity in a gene dosage dependent manner
We next utilized catalytically-inactive CRISPR/dCas9 (referred to as CRISPRi)(26) to knockdown (KD) but not completely eliminate CDK12 expression. We tested the silencing efficiency of multiple sgRNAs targeting CDK12 to generate stable KDs (Supplementary Fig. S3A) and chose the most robust sgRNA (sgRNA-2) to express in multiple prostate cancer (e.g., LNCaP and PC3 cells) and a colon cancer (DLD-1) cell line. We used ovarian cancer (OVCAR8 and CAOV3) and breast cancer (MDA-MB-231) cell lines as controls. In all cancer types tested, we found that silencing CDK12 led to PARPi sensitivity, nearly to the same degree as silencing BRCA2 (Fig. 2A–2E, Supplementary Fig. S3B), suggesting that the synthetic lethal relationship between CDK12 and PARPi is observed across multiple tumor cell types. In addition, in prostate cancer lines, PARPi sensitivity was not dependent on expression of androgen receptor (AR), since CDK12 loss in PC3 cells (AR-negative, Fig. 2B), TRAMP-C2 (AR-negative, Supplementary Fig. S2F–S2G), LNCaP (AR-positive) and C42B cells (AR-positive) all showed enhanced sensitivity to PARPi. In the CRISPRi cells, the degree of CDK12 expression inhibition was approximately 70% as measured by western blotting and quantitative PCR (qPCR) (Fig. 2F–2G). Interestingly, when comparing the degree of PARPi sensitization in the LNCaP CDK12 KD versus KO cells, we noticed more pronounced differences in PARPi sensitivity in the complete KO cells compared to the KD and control cells (Fig. 2H). This was also seen with BRCA2, in which DLD1 cells with a complete BRCA2 KO were more sensitive to PARPi than BRCA2 KD cells (Supplementary Fig. S3C). Cells in which only one allele of CDK12 was edited by CRISPR/Cas9 (i.e. a monoallelic deletion), resulting in at least one remaining intact, wildtype, allele of CDK12, were not more sensitive to PARPi, although levels of CDK12 were mildly reduced (Fig. 2I–2J). Taken together, these data demonstrate that CDK12 loss in multiple cancer cell types leads to PARPi sensitivity, which is dependent on the residual gene dosage of CDK12; cells with complete CDK12 KO are more sensitive to PARPi than cells with partial CDK12 loss.
Figure 2: CDK12 loss in multiple cancer cell types results in PARPi sensitivity in a gene dosage dependent manner and CDK12 is necessary for HR repair and gene expression.
(A-E) Dose response assays to talazoparib using CRISPRi to knockdown CDK12 or BRCA2 in LNCaP (A), PC3 (B), CAOV3 (C), OVCAR8 (D), and DLD-1 (E) cells. A representative assay of n=4 independent experiments performed in technical triplicates is shown for each cell line. (F) WB showing protein levels of CDK12 in control and CRISPRi knockdown in LNCaP cells. (G) mRNA levels of CDK12 and BRCA2 in control and CRISPRi knockdown in LNCaP cells, as measured by QPCR. **p<0.01, n=3 independent samples. (H) Dose response assay to talazoparib in LNCaP CDK12 KO versus CDK12 CRISPRi KD cells. (I) Dose response assays to olaparib in LNCaP CDK12 heterozygous (Het1 and Het2) versus KO cells, compared to controls. (J) WB showing protein levels of CDK12 levels in control, KO, Het1 and Het2 cells (K) U2OS-DR-GFP reporter cells were transfected with ISce-I and siRNAs targeting CDK12 or BRCA2 and GFP fluorescence was measured. ***p<0.001, n=3 independent assays performed in triplicate. (L) U2OS-DR-GFP reporter cells were treated with increasing doses of THZ531 (0–400nM), transfected with ISce-I and GFP fluorescence was measured. *p<005, **p<0.01, n=3 independent assays performed in triplicate. (M) Quantification of comet tail:head DNA ratios in C42B control and CDK12 KO cells treated with or without 1μM rucaparib for 5 days. At least 100 nuclei were counted for each cell line and condition. Box and whiskers plot shows a representative experiment of n=3 independent experiments. ns=not significant, ***p<0.001. (N) mRNA levels of indicated DNA repair genes in C42B cells treated with 250 nM of THZ531, as measured by QPCR (n=5 samples at each concentration, graph depicts mean SEM, *p<0.05, **p<0.01, ***p<0.001, p<0.0001, ns=not significant). Error bars represent SEM in A-E, G-I, and K-N. Student’s t test was performed for G. ANOVA with multiple comparison correction (Sidak) was performed for K-N.
CDK12 is necessary for homologous recombination repair activity and gene expression
We next tested the functional significance of CDK12 loss on homologous recombination (HR) repair activity. We silenced CDK12 in the U2OS-DR-GFP cells, which carry a HR reporter (27), and found that CDK12 inhibition decreased reporter fluorescence after I-SceI transfection, nearly to the same degree as BRCA2 inhibition (Fig. 2K). Interestingly, treating U2OS-DR-GFP cells with THZ531, a covalent CDK12/13 inhibitor (28), also suppressed HR reporter activity in a dose-dependent manner (Fig. 2L). At baseline, we did not detect more endogenous double-stranded breaks in CDK12 KO cells. However, PARPi treatment specifically increased double-stranded breaks in the CDK12 KO cells, as measured by comet assays (Fig. 2M). In accordance with prior studies showing an enrichment of DNA repair pathway genes affected in the setting of CDK12 inhibition (17–19,29–31), treating C42B PC cells with THZ531 (250 nM) also suppressed expression of multiple genes involved in DNA repair, including BRCA1 and BRCA2 (Fig. 2N). Interestingly, BRCA1, BRCA2 and other DNA repair genes were not consistently downregulated in the CDK12 KO cells, suggesting possible compensatory mechanisms to restore their expression in the chronic versus acute CDK12 deficient conditions (Supplementary Fig. S3D–S3E). Taken together, these results demonstrate that CDK12 is necessary for optimal HR activity and that pharmacologic inhibition, but not chronic loss, of CDK12 affects the expression of multiple DNA repair genes.
Loss of cyclin K, but not CDK13, impairs HR and results in PARPi sensitivity
The kinase domains of CDK12 and CDK13 are 92% identical, and both CDK12 and CDK13 bind to same cognate cyclin, cyclin K (32). Although CDK12 and CDK13 have been shown to have distinct functions (33), both depend on interactions with cyclin K to execute their kinase functions (14). We silenced cyclin K (CCNK) and CDK13 using CRISPRi (Fig. 3A) and found that inhibition of cyclin K, but not CDK13, conferred PARPi sensitivity (Fig. 3B). In addition, we generated single-cell derived CDK13 KO clones (Fig.3C) and found that in contrast to the CDK12 KO cells, the CDK13 KO cells were not more sensitive to PARPi and behaved similarly to control cell lines (Fig. 3D). Accordingly, silencing CCNK but not CDK13 suppressed HR reporter activity (Fig. 3E). Taken together, these data demonstrate that despite highly similar kinase domains, CDK13 does not affect HR activity or PARPi sensitivity, suggesting that the CDK12-Cyclin K heterodimer is specifically critical for these functions. Interestingly, mutations and deletions of cyclin K are found in multiple cancer types, including cervical, endometrial and esophageal squamous cell carcinoma (Supplementary Fig. S4), suggesting that additional tumor types may also been sensitive to PARPi.
Figure 3: Loss of cyclin K (CCNK) but not CDK13 impairs HR and results in PARPi sensitivity.
(A) mRNA levels of CCNK and CDK13 in C42B CRISPRi cells expressing an sgRNA targeting CCNK or CDK13 (n=3 samples, graph depicts mean SEM, **p<0.01). (B) Dose response assay to rucaparib in C42B CRISPRi cells targeting CCNK or CDK13. (C) WB of CDK13 in C42B CDK13 KO cells. GAPDH is shown as loading control. (D) Dose response assay to rucaparib in control and CDK13 KO cells. A representative assay of n=3 independent experiments performed in technical triplicates is shown. (E) U2OS-DR-GFP reporter cells were transfected with ISce-I and siRNAs targeting CDK13, CCNK, CDK12 or BRCA2 and GFP fluorescence was measured. A representative assay of n=3 independent experiments performed in technical triplicates is shown. Error bars represent SEM in A-B, D-E. Student’s t test was performed for A. ANOVA with multiple comparison correction (Sidak) was performed for E.
CDK12 kinase domain and truncation mutants differentially affect PARPi sensitivity
Given the importance of the kinase domain, we next asked whether two recurrent kinase domain point mutations identified in PC patients (R858W and D918G, which are labeled as variants of unknown significance (VUS) on cBioPortal) altered PARPi sensitivity (Fig. 4A). The R858 residue is one of three critical arginine residues located within the active kinase center that stabilizes the T-loop upon phosphorylation of a critical threonine residue T893 (34,35). To test the function of these mutants, we constitutively expressed wildtype (WT) CDK12, and the R858W and D918G kinase domain point mutants in LNCaP CDK12 KO cells (Fig. 4B). Expression of LacZ was used a control. While expression of CDK12 WT (magenta) completely restored PARPi resistance, expression of the R858W (yellow) and D918G (green) point mutants (which likely have reduced kinase activity) resulted in a partial rescue of PARPi sensitivity from an IC50 of 0.003 μM to an IC50 of 0.05–0.10 μM (Fig. 4C). In addition, the R858W and D918G point mutants partially rescued PARPi sensitivity in the C42B CDK12 KO cells (Supplementary Fig. S5A).
Figure 4: Kinase domain and truncation mutants differentially affect PARPi sensitivity.
(A) Lollipop diagram showing kinase domain point mutations in CDK12, including the two more frequently occurring mutations, R858W and D918G. (B) WB showing expression of LacZ, CDK12 WT, CDK12 R858W and CDK12 D918G in LNCaP control and CDK12 KO cells. GAPDH is shown as the loading control. All proteins were generated with an amino terminus V5 tag. (C) Dose response assay to rucaparib in LNCaP control and CDK12 KO cells expressing WT, R858W or D918G point mutants. (D) Schematic of full-length CDK12 and truncation mutants (KDΔ, PRMΔ, and RSΔ). The number to right represents the last amino acid of full-length CDK12 and each mutant. (E) WB of expression of truncation mutants in LNCaP cells. GAPDH is shown the loading control. (F) Dose response assay to rucaparib in LNCaP control and CDK12 KO cells expressing WT, KDΔ, PRMΔ, and RSΔ truncation mutants. (G) Immunoprecipitation of doxycycline-inducible FLAG-tagged WT, KDΔ, PRMΔ, and RSΔ mutants. Eluate was blotted for FLAG and endogenous cyclin K. The input for cyclin K is shown. For panels C and F, a representative assay of n=3 independent experiments performed in technical triplicates is shown. Error bars represent SEM in C and F.
Next, we expressed truncated forms of CDK12, which are commonly found in prostate cancer patients (12), and made deletions C-terminal to the known functional protein domains: RSΔ (amino acids 1 – 386), PRMΔ (amino acids 1 – 700) and kinase domain (KDΔ) (amino acids 1 – 1020) (Fig. 4D–4E). None of the truncation mutants were sufficient to fully restore PARPi sensitivity in LNCaP or C42B CDK12 KO cells, with IC50 ranges of 0.002 – 0.004 μM compared to the WT, which shifted the IC50 to 0.3 μM (Fig. 4F, Supplementary Fig. S5B). These data suggest that not only is the kinase domain important in mediating PARPi sensitivity, but that the carboxy terminal of CDK12 beyond the kinase domain is also critical. Indeed, prior studies demonstrated that a carboxy terminal extension from the CDK12 kinase domain which is located between amino acids 1044–1063 was important for kinase activity (34). Interestingly, the majority (96%, 262/271 examined in cBioportal) of truncating mutations in PC reported occur prior to the carboxy terminal extension (amino acid 1063), further suggesting the importance of the carboxy terminal extension. The phosphorylation levels of serine 2 (Ser2) RNA Pol II were slightly reduced or similar in the CDK12 KO, point mutants or truncation mutants cells compared to control, while levels of serine 5 (Ser5) and serine 7 (Ser7) phosphorylation were overall similar (Supplementary Fig. S5C–5D). Finally, we asked whether the truncated forms of CDK12 were able to interact with cyclin K, since the interaction with cyclin K is critical for CDK12 kinase activity. Co-immunoprecipitation (co-IP) experiments using doxycycline-inducible FLAG-tagged CDK12 WT, KDΔ, PRMΔ and RSΔ constructs demonstrated that none of the truncated mutants were able to interact with cyclin K (Fig. 4G). Taken together, these data demonstrate that CDK12 kinase domain mutants and truncation mutants are unable to fully restore PARPi sensitivity. In addition, CDK12 truncation mutants cannot interact with cyclin K, which is a critical partner for the kinase function of CDK12, although there may also be additional unknown kinase-independent functions of CDK12.
CDK12 KO prostate cancer xenografts are sensitive to PARPi monotherapy
We next asked whether CDK12 KO tumor xenografts were sensitive to PARPi monotherapy in vivo. Rucaparib treatment inhibited tumor growth in C42B CDK12 KO xenografts, whereas treatment of control C42B xenografts showed slight growth inhibition that was not statistically significant (Fig. 5A–5B). We confirmed that tumors explanted at the end of the experiment remained null for CDK12 by western blot and immunohistochemistry (Fig. 5C–5D). CDK12 KO tumors also had only slightly reduced levels of phosphorylated Ser2-RNA Pol II (Fig. 5C). Rucaparib treatment also increased γH2AX staining (marker of double-strand DNA breaks) in the CDK12 KO, but not control CDK12 WT tumors and decreased phospho-histone H3 staining, a marker of proliferation, consistent with enhanced sensitivity to PARPi in CDK12 KO tumors in vivo (Fig. 5E–5F). Taken together, our data demonstrate that PARPi monotherapy inhibits CDK12 KO tumor growth in vivo, and is associated with increased markers of DNA damage and decreased proliferation.
Figure 5: CDK12 KO prostate cancer xenografts are more sensitive to PARPi monotherapy.
(A) Tumor growth measurements in C42B control and CDK12 KO tumor xenografts, treated with or without rucaparib (150 mg/kg daily). n = 8 mice per group, graph depicts mean tumor size SEM. * denotes p<0.05 and ** denotes p<0.01. (B) The mean percentage change of tumor volume for C42B control and CDK12 KO tumor xenografts, treated with or without rucaparib. (C) WB of tumor xenografts for CDK12 and phosphoSer2-RNA Pol II. (D) Immunohistochemical images for CDK12 in control and CDK12 KO tumor xenografts after rucaparib treatment. Scale bar = 100μm. (E-F) Representative images of γH2AX (E) and phospho-histone H3 (F) immunohistochemical images and quantification (right). n=4 fields per tumor, 5 tumors per cohort, ns=not significant, **p<0.01, ****p<0.001. Errors bars represent SEM in A-B. Horizontal bars represent the mean in E-F. ANOVA with multiple comparison correction (Sidak) was performed for A-B and E-F.
Patients with CDK12-mutated prostate cancer exhibit responses to rucaparib monotherapy
Finally, we asked whether prostate cancer patients with CDK12 alterations had any clinical benefit from PARPi monotherapy. TRITON2 (NCT02952534) was a phase 2 clinical trial evaluating rucaparib monotherapy in patients with advanced castrate-resistant prostate cancer (CRPC) who progressed after receiving an androgen pathway inhibitor (e.g., enzalutamide or abiraterone) and taxane chemotherapy (23). A total of 15 patients were included in the CDK12 cohort (Supplementary Table S2) (24). We found that 11 patients had biallelic CDK12 alterations (which included two patients with a single detectable CDK12 mutation but had evidence of loss of heterozygosity (LOH) of the second wildtype allele based on sequencing). Of the biallelic mutated patients, 7 of the 11 patients (63.6%) had a reduction in serum prostate specific antigen (PSA) levels from baseline (range −1.4% to 74.9%, Fig. 6A), and 5 of the 11 patients (45.4%) had a PSA30 response or better (representing a PSA reduction of 30% or more from baseline after starting rucaparib). Of the patients with biallelic CDK12 alterations and evaluable lesions on computed tomography (CT) scans, 4 of 6 patients (66.6%) had radiographic disease control, with stable disease (SD) as their best response per RECIST (response evaluation criteria in solid tumors) (Fig. 6B). We also analyzed the PSA trends for individual patients who responded to rucaparib. Patient 1 had a greater than PSA50 response and the PSA remained below the initial baseline for 32 weeks (Fig. 6C). Meanwhile, Patient 2 had a less than PSA50 response, but the PSA remained below the initial baseline for 39 weeks (Fig. 6D). Although these responses do not meet the clinical response criteria or primary objectives pre-specified in the study design of TRITON2, the data suggest that rucaparib monotherapy may have activity in heavily pre-treated patients with CDK12 alterations, who would otherwise have few remaining therapeutic options at this advanced stage of disease. Interestingly, one patient who had a single CDK12 mutation (whom we classified as monoallelic, since only one mutation was detected and there was no evidence of LOH of the second allele) did not have a PSA response. In addition, 0 of 3 patients with a CDK12 gene arrangement (with indeterminate zygosity) had a PSA30 response (Fig. 6A, Supplementary Table S2). Taken together, these data demonstrate that PARP inhibitors may have modest anti-tumor activity in advanced CRPC patients with CDK12 alterations, with responses observed only in the subset of patients with biallelic alterations.
Figure 6: Responses to rucaparib in prostate cancer patients varies by the type of CDK12 mutation.
(A) Waterfall plot of best percent change in serum prostate specific antigen (PSA) levels in patients with monoallelic (light shaded gray), gene arrangements (dark gray) or biallelic (black) CDK12 mutations treated with rucaparib monotherapy on the TRITON2 trial. (B) Four of 6 patients with measurable disease showed stable disease (SD), while 2 of 6 patients showed progressive disease (PD) by RECIST. (C-D) PSA values in patient 1 who achieved a PSA50 response (C) and patient 2 who did not achieve a PSA50 response (D) treated with rucaparib monotherapy on the TRITON2 trial. The baseline PSA value for each patient is indicated by the dotted line.
DISCUSSION
CDK12 mutations are found in approximately 5–7% of mCRPC and 3% of ovarian cancer patients (12,36,37). Metastatic PC patients with CDK12 mutated tumors have multiple features of aggressive disease, including a shorter time to metastasis and shorter time to castration resistance (12). Here, we used CRISPRn and CRISPRi to generate pre-clinical PC cell line models of CDK12 deficiency and performed a small molecule screen of 1800 compounds to identify synthetic lethal interactions. Amongst our top hits, multiple PARPi’s were selectively lethal to CDK12-deficient cells. We further discovered that complete loss of CDK12 conferred the most sensitivity to PARPi-mediated death; cells with suppressed but still detectable amounts of CDK12 displayed intermediate sensitivity, which has important implications for patient selection in CDK12 specific clinical trials. In addition, the synthetic lethal relationship between CDK12 loss and PARPi was observed across multiple tumor types that were evaluated. This is important to establish since targeting genetic alterations in one cancer type does not necessarily generalize to other cancers, and basket trials may mask the effect of a promising drug target. For example, BRAF inhibitors targeting the V600E mutations demonstrate considerable activity in melanoma but not in colorectal and other cancers (38–40). Furthermore, while some studies have not shown PARPi sensitivity in CDK12-deficient ovarian cancer cells (41), our results using multiple inactivation methods across multiple tumor cell lines, including ovarian cancer, indicate that CDK12 loss may be broadly targetable using PARPi, potentially impacting a wide range of cancers characterized by CDK12 loss, including ovarian, endometrial, gastroesophageal and others (42). This hypothesis warrants further investigation, given recent findings that the degree of PARPi sensitivity across tumor histology varies even across BRCA1/2 mutated cancers, likely depending on whether mutant BRCA1/2 is a driver or bystander mutation (43).
Our data show that CCNK inhibition also leads to PARPi sensitivity, in accordance with a previous study(44), suggesting that additional molecular subsets of cancer with mutated or reduced CCNK expression may be treated with PARPi. Analysis of TCGA and cBioportal data demonstrated that esophageal, cervical and endometrial carcinomas, as well as other tumor types, may harbor CCNK mutations and deletions. Interestingly, CCNK downregulation may be a feature of enzalutamide resistance and targetable by PARPi(44). Furthermore, despite being over 92% similar in their kinase domains, loss of CDK13 did not confer PARPi sensitivity. This is consistent with previous studies showing that these two CDKs regulate different cellular processes (18,33), though the molecular basis for this distinction remains unclear. Interestingly, both CDK12/13 kinase inhibitors and CDK12 degraders have been shown to increase PARPi sensitivity in preclinical models of breast cancer and myeloma (30,31,45–48).
We provide multiple lines of in vivo evidence to support anti-tumor activity of PARPi in bona fide CDK12-deficient prostate tumors. Mice bearing CDK12 KO tumor xenografts were sensitive to rucaparib monotherapy, which significantly repressed tumor growth and induced double strand DNA breaks in vivo. We also showed that in the TRITON2 study, rucaparib had moderate monotherapy activity in the subset of patients with biallelic CDK12 mutations. Although few patients in the CDK12 cohort had PSA50 responses (21), we found that rucaparib monotherapy elicited PSA reductions in 63.6% of patients with biallelic CDK12 mutations, with 45.4% of patients achieving a PSA30 or better response. The trial is limited by the number of CDK12 patients. While these data do not meet the clinical PSA50 criteria nor the prespecified threshold for primary objective response in the trial, they do indicate that in this heavily pretreated patient population with limited remaining therapeutic options, PARPi therapy may have some activity to stabilize disease and underscore the importance of selecting patients with biallelic loss or mutations. While the mechanism of PARPi response in the context of CDK12 deficiency remains unclear, this may be partly attributable to CDK12’s ability to regulate expression of HR repair genes (17,18,28,29), although additional mechanisms may exist. For example, whether transcription-replication conflicts, which are increased in CDK12 KO cells (49), may contribute to the PARPi sensitivity phenotype (50) remains to be explored.
Recently, a pooled analysis of multiple mCRPC trials investigating PARPi monotherapy or in combination with an androgen receptor pathway inhibitor (ARPI) was performed, and evaluated the contribution of adding PARPi by HRR gene mutation. This study found that in CDK12-mutated patients, the radiographic progression free survival (rPFS) hazard ratio (HR) was 0.50 (confidence interval, 0.32 to 0.80) and the overall survival (OS) HR was 0.63 (confidence interval, 0.29 to 0.99)(51). These benefits are somewhat comparable to BRCA2-mutated patients, which showed an rPFS HR of 0.31 (confidence interval, 0.23 to 0.42) and OS HR of 0.66 (confidence interval, 0.49 to 0.89)(51). The pooled analysis included data from the PROpel (52), PROfound (53,54), TALAPRO1 (55), TALAPRO2 (22), MAGNITUDE (56), and TRITON2 (23,24,57) trials. Whether there are mechanistic differences with combining PARPi with direct AR antagonists (such as enzalutamide) versus adrenal androgen synthesis inhibitors (such as abiraterone) require further study. However, the studies to date suggest that the combination with abiraterone may be less beneficial(52,58) compared to the combination with enzalutamide in CDK12-mutated patients (HR of 0.49, confidence interval 0.23 to 1.02)(59). The clinical trial results are consistent with our findings here, and we propose that PARPi’s should be incorporated in combination regimens for patients that harbor biallelic CDK12 mutations. Patients with a monoallelic CDK12 mutation (without evidence for LOH of the second allele) or gene rearrangements may be less likely to respond to PARPi, as they may still have residual CDK12 expression, and thus should be excluded. Although there was only one patient with a monoallelic CDK12 mutation in the TRITON2 trial that we could verify, this patient did not respond to rucaparib. The heterogeneous and/or monoallelic CDK12 mutations found in PC may explain why prior studies with limited samples sizes failed to show benefit with PARPi in men with CDK12 mutant PC (24,60).
Our study also elucidates how the nature of the mutation affects CDK12 function and therapeutic response. Most patients enrolled on “CDK12 selected” trials are generally eligible regardless of where the frameshift or point mutation occurs in the gene. We showed that shorter truncation mutants found in PCa patients are unable to bind to cyclin K, the requisite binding partner of CDK12 important for its catalytic kinase function and critical for PARPi sensitivity. In addition, given the other protein domains found in CDK12 (such as the arginine-serine rich (RS) and proline-rich motif (PRM), CDK12 may have additional, kinase-independent functions, which will also be important to understand. While the majority of patient CDK12 mutations are frameshift mutations leading to early truncations, a subset of prostate and ovarian tumors harbor kinase domain point mutations (12,61). We found that kinase domain mutants with partial kinase function showed intermediate PARPi sensitivity, whereas truncation mutants without any residual kinase function behaved similarly to the KO cells and retained full sensitivity to PARPi. Interestingly, of the eight CDK12 patients on TRITON2 who had reductions in their serum PSA, the top four patients had two CDK12 mutations that each led to early truncations before the kinase domain; the next 2 patients with PSA reductions each had one kinase domain mutation and one truncation mutation, and showed an intermediate response; and the two patients with a single detected mutation and predicted LOH of the second allele (and therefore we classified as biallelic) had no PSA response. These data underscore the notion that patients with different CDK12 alterations (i.e., early frameshift truncations versus kinase domain point mutations, and biallelic mutations versus biallelic through LOH) may need different therapeutic approaches, and that combinations may be required to increase clinical benefit for certain types of CDK12 alterations. Knowing specific genetic alterations will therefore be important in selecting patients and identifying robust, predictive biomarkers. We await additional evidence to support this hypothesis, which will likely emerge from completed and other ongoing clinical trials, and advocate for greater transparency that trials report patient-level specific mutations and response rates.
Finally, our chemical screen in isogenic CDK12 deficient cells also identified other potential synthetic lethal drugs, which we plan to test in future studies. Recent studies also suggest other approaches to target CDK12 deficiency, including suppressing CDK13 (62) and WEE1 (49). Prior work from our group and others showed that CDK12-mutant prostate cancers had high numbers of T cell immune infiltrates and elevated neoantigen fusions (36,63), although a clinical trial to evaluate checkpoint immunotherapy in CDK12-mutant mCRPC showed poor responses (64). Additional work to target this aggressive subtype of prostate cancer is urgently needed.
In conclusion, our data support continued investigations of PARPi in patients with biallelic CDK12 alterations. Our data also point to the need to further understand the biological significance of different CDK12 mutations found in cancer patients and caution against broadly grouping patients with CDK12 alterations into trials evaluating the efficacy of PARPi and potentially other targeted agents.
Supplementary Material
STATEMENT OF TRANSLATIONAL RELEVANCE.
CDK12-deficient cancer cells are sensitive to PARP inhibitor (PARPi) in a gene-dosage and mutation dependent manner. Loss of cyclin K, but not CDK13, also leads to PARPi sensitivity. Kinase domain and truncation mutations in CDK12 lead to differential sensitivities to PARPi. In the TRITON2 clinical trial evaluating the efficacy of rucaparib monotherapy in advanced metastatic castration-resistant prostate cancer (mCRPC) patients, more than 50% of mCRPC patients with bona fide, biallelic mutations in CDK12 had reductions in their serum PSA. Our findings support further investigation of PARPi synthetic lethality in biallelic, CDK12-mutated advanced prostate cancer.
ACKNOWLEDGEMENTS
We thank members of the Feng and Ashworth Laboratories for feedback and critical reading of the manuscript. We thank Dr. Bradley Stohr (UCSF) for helpful discussions, the UCSF Laboratory for Cell Analysis (J. Gordon and S. Elmes) for assistance with flow cytometry, and Dr. Grace Cheng (University of British Columbia) and Dr. Matija Peterlin (UCSF) for CDK12 constructs.
We acknowledge and thank pharmaand Gmbh (pharma&) as the new owner of rucaparib since this research was conducted.
J.Chou has been supported by a postdoctoral fellowship from the A.P. Giannini Foundation, a training grant from the National Cancer Institute (T32 CA108462), the Department of Defense Prostate Cancer Research Program Physician Research Award (W81XWH-20-1-0136), and a Prostate Cancer Foundation Young Investigator Award.
A.M. Chinnaiyan and F.Y. Feng are supported by a SPORE grant (1P50CA186786).
D.A. Quigley is supported by Young Investigator awards from the Prostate Cancer Foundation and the BRCA Foundation.
F.Y. Feng and A. Ashworth are supported by Challenge Awards from the Prostate Cancer Foundation, the Benioff Institute for Prostate Cancer Research and the National Cancer Institute (1R01CA230516).
Abbreviations:
- CDK12
Cyclin Dependent Kinase-12
- PARP
poly (ADP-ribose) polymerase inhibitor
- PC
prostate cancer
Footnotes
CONFLICTS OF INTEREST
W.A. reports honoraria from Roche, Medscape, Aptitude Health, Clinical Education Alliance, OncLive/MJH Life Sciences, touchIME, and Pfizer; serves in a consulting or advisory role for Clovis Oncology, Janssen, ORIC Pharmaceuticals, Daiichi Sankyo and AstraZeneca/MedImmune; and reports research funding from AstraZeneca, Zenith Epigenetics, Clovis Oncology, ORIC Pharmaceuticals, Epizyme, and Nuvation Bio to his institution.
J.C. serves as a consultant for ExaiBio.
M.N., A.L., T.G. and A.S. were former employees at Clovis Oncology.
A.M.C. is a co-founder of and serves as a Scientific Advisory Board member for LynxDx, Esanik Therapeutics, Medsyn, and Flamingo Therapeutics; he is a scientific advisor or consultant for EdenRoc, Proteovant, Belharra, Rappta Therapeutics, and Tempus.
F.Y.F. is currently serving or has served on the advisory boards or has received consulting fees from Astellas, Bayer, Celgene, Clovis Oncology, Janssen, Genentech Roche, Myovant, Roivant, Sanofi and Blue Earth Diagnostics; he also is a member of the SAB for Artera, ClearNote Genomics, SerImmune, and BMS (Microenvironment Division).
A.A. is a co-founder of Tango Therapeutics, Azkarra Therapeutics, Ovibio Corporation and Kytarro, a member of the board of Cytomx and Cambridge Science Corporation, a member of the scientific advisory board of Genentech, GLAdiator, Circle, Bluestar, Earli, Ambagon, Phoenix Molecular Designs, Yingli, Oric, Hap10 and Trial Library, a consultant for SPARC, ProLynx, Novartis and GSK, receives research support from SPARC, and holds patents on the use of PARP inhibitors held jointly with AstraZeneca from which he has benefited financially (and may do so in the future).
All other authors report no relevant disclosures.
REFERENCES
- 1.Ramalingam SS, Vansteenkiste J, Planchard D, Cho BC, Gray JE, Ohe Y, et al. Overall Survival with Osimertinib in Untreated, EGFR-Mutated Advanced NSCLC. N Engl J Med 2020;382(1):41–50 doi 10.1056/NEJMoa1913662. [DOI] [PubMed] [Google Scholar]
- 2.Drilon A, Laetsch TW, Kummar S, DuBois SG, Lassen UN, Demetri GD, et al. Efficacy of Larotrectinib in TRK Fusion-Positive Cancers in Adults and Children. N Engl J Med 2018;378(8):731–9 doi 10.1056/NEJMoa1714448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Lord CJ, Ashworth A. PARP inhibitors: Synthetic lethality in the clinic. Science 2017;355(6330):1152–8 doi 10.1126/science.aam7344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Golan T, Hammel P, Reni M, Van Cutsem E, Macarulla T, Hall MJ, et al. Maintenance Olaparib for Germline BRCA-Mutated Metastatic Pancreatic Cancer. N Engl J Med 2019;381(4):317–27 doi 10.1056/NEJMoa1903387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Coleman RL, Oza AM, Lorusso D, Aghajanian C, Oaknin A, Dean A, et al. Rucaparib maintenance treatment for recurrent ovarian carcinoma after response to platinum therapy (ARIEL3): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017;390(10106):1949–61 doi 10.1016/S0140-6736(17)32440-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Litton JK, Rugo HS, Ettl J, Hurvitz SA, Goncalves A, Lee KH, et al. Talazoparib in Patients with Advanced Breast Cancer and a Germline BRCA Mutation. N Engl J Med 2018;379(8):753–63 doi 10.1056/NEJMoa1802905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Mateo J, Carreira S, Sandhu S, Miranda S, Mossop H, Perez-Lopez R, et al. DNA-Repair Defects and Olaparib in Metastatic Prostate Cancer. N Engl J Med 2015;373(18):1697–708 doi 10.1056/NEJMoa1506859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.McCabe N, Turner NC, Lord CJ, Kluzek K, Bialkowska A, Swift S, et al. Deficiency in the repair of DNA damage by homologous recombination and sensitivity to poly(ADP-ribose) polymerase inhibition. Cancer Res 2006;66(16):8109–15 doi 10.1158/0008-5472.CAN-06-0140. [DOI] [PubMed] [Google Scholar]
- 9.Robinson D, Van Allen EM, Wu YM, Schultz N, Lonigro RJ, Mosquera JM, et al. Integrative Clinical Genomics of Advanced Prostate Cancer. Cell 2015;162(2):454 doi 10.1016/j.cell.2015.06.053. [DOI] [PubMed] [Google Scholar]
- 10.Armenia J, Wankowicz SAM, Liu D, Gao J, Kundra R, Reznik E, et al. The long tail of oncogenic drivers in prostate cancer. Nat Genet 2018;50(5):645–51 doi 10.1038/s41588-018-0078-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Quigley DA, Dang HX, Zhao SG, Lloyd P, Aggarwal R, Alumkal JJ, et al. Genomic Hallmarks and Structural Variation in Metastatic Prostate Cancer. Cell 2018;174(3):758–69 e9 doi 10.1016/j.cell.2018.06.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Reimers MA, Yip SM, Zhang L, Cieslik M, Dhawan M, Montgomery B, et al. Clinical Outcomes in Cyclin-dependent Kinase 12 Mutant Advanced Prostate Cancer. Eur Urol 2020;77(3):333–41 doi 10.1016/j.eururo.2019.09.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Nguyen B, Mota JM, Nandakumar S, Stopsack KH, Weg E, Rathkopf D, et al. Pan-cancer Analysis of CDK12 Alterations Identifies a Subset of Prostate Cancers with Distinct Genomic and Clinical Characteristics. Eur Urol 2020;78(5):671–9 doi 10.1016/j.eururo.2020.03.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Cheng SW, Kuzyk MA, Moradian A, Ichu TA, Chang VC, Tien JF, et al. Interaction of cyclin-dependent kinase 12/CrkRS with cyclin K1 is required for the phosphorylation of the C-terminal domain of RNA polymerase II. Mol Cell Biol 2012;32(22):4691–704 doi 10.1128/MCB.06267-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Bartkowiak B, Liu P, Phatnani HP, Fuda NJ, Cooper JJ, Price DH, et al. CDK12 is a transcription elongation-associated CTD kinase, the metazoan ortholog of yeast Ctk1. Genes Dev 2010;24(20):2303–16 doi 10.1101/gad.1968210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Tien JF, Mazloomian A, Cheng SG, Hughes CS, Chow CCT, Canapi LT, et al. CDK12 regulates alternative last exon mRNA splicing and promotes breast cancer cell invasion. Nucleic Acids Res 2017;45(11):6698–716 doi 10.1093/nar/gkx187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Dubbury SJ, Boutz PL, Sharp PA. CDK12 regulates DNA repair genes by suppressing intronic polyadenylation. Nature 2018;564(7734):141–5 doi 10.1038/s41586-018-0758-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Blazek D, Kohoutek J, Bartholomeeusen K, Johansen E, Hulinkova P, Luo Z, et al. The Cyclin K/Cdk12 complex maintains genomic stability via regulation of expression of DNA damage response genes. Genes Dev 2011;25(20):2158–72 doi 10.1101/gad.16962311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Bajrami I, Frankum JR, Konde A, Miller RE, Rehman FL, Brough R, et al. Genome-wide profiling of genetic synthetic lethality identifies CDK12 as a novel determinant of PARP1/2 inhibitor sensitivity. Cancer Res 2014;74(1):287–97 doi 10.1158/0008-5472.CAN-13-2541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Joshi PM, Sutor SL, Huntoon CJ, Karnitz LM. Ovarian cancer-associated mutations disable catalytic activity of CDK12, a kinase that promotes homologous recombination repair and resistance to cisplatin and poly(ADP-ribose) polymerase inhibitors. J Biol Chem 2014;289(13):9247–53 doi 10.1074/jbc.M114.551143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Abida W, Campbell D, Patnaik A, Shapiro JD, Sautois B, Vogelzang NJ, et al. Non-BRCA DNA Damage Repair Gene Alterations and Response to the PARP Inhibitor Rucaparib in Metastatic Castration-Resistant Prostate Cancer: analysis from the phase 2 TRITON2 study. Clin Cancer Res 2020. doi 10.1158/1078-0432.CCR-20-0394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Agarwal N, Azad AA, Carles J, Fay AP, Matsubara N, Heinrich D, et al. Talazoparib plus enzalutamide in men with first-line metastatic castration-resistant prostate cancer (TALAPRO-2): a randomised, placebo-controlled, phase 3 trial. Lancet 2023;402(10398):291–303 doi 10.1016/S0140-6736(23)01055-3. [DOI] [PubMed] [Google Scholar]
- 23.Abida W, Patnaik A, Campbell D, Shapiro J, Bryce AH, McDermott R, et al. Rucaparib in Men With Metastatic Castration-Resistant Prostate Cancer Harboring a BRCA1 or BRCA2 Gene Alteration. J Clin Oncol 2020;38(32):3763–72 doi 10.1200/JCO.20.01035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Abida W, Campbell D, Patnaik A, Shapiro JD, Sautois B, Vogelzang NJ, et al. Non-BRCA DNA Damage Repair Gene Alterations and Response to the PARP Inhibitor Rucaparib in Metastatic Castration-Resistant Prostate Cancer: Analysis From the Phase II TRITON2 Study. Clin Cancer Res 2020;26(11):2487–96 doi 10.1158/1078-0432.CCR-20-0394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Chou J, Quigley DA, Robinson TM, Feng FY, Ashworth A. Transcription-Associated Cyclin-Dependent Kinases as Targets and Biomarkers for Cancer Therapy. Cancer Discov 2020;10(3):351–70 doi 10.1158/2159-8290.CD-19-0528. [DOI] [PubMed] [Google Scholar]
- 26.Qi LS, Larson MH, Gilbert LA, Doudna JA, Weissman JS, Arkin AP, et al. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 2013;152(5):1173–83 doi 10.1016/j.cell.2013.02.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Pierce AJ, Johnson RD, Thompson LH, Jasin M. XRCC3 promotes homology-directed repair of DNA damage in mammalian cells. Genes Dev 1999;13(20):2633–8 doi 10.1101/gad.13.20.2633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Zhang T, Kwiatkowski N, Olson CM, Dixon-Clarke SE, Abraham BJ, Greifenberg AK, et al. Covalent targeting of remote cysteine residues to develop CDK12 and CDK13 inhibitors. Nat Chem Biol 2016;12(10):876–84 doi 10.1038/nchembio.2166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Krajewska M, Dries R, Grassetti AV, Dust S, Gao Y, Huang H, et al. CDK12 loss in cancer cells affects DNA damage response genes through premature cleavage and polyadenylation. Nat Commun 2019;10(1):1757 doi 10.1038/s41467-019-09703-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Jiang B, Gao Y, Che J, Lu W, Kaltheuner IH, Dries R, et al. Discovery and resistance mechanism of a selective CDK12 degrader. Nat Chem Biol 2021;17(6):675–83 doi 10.1038/s41589-021-00765-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Quereda V, Bayle S, Vena F, Frydman SM, Monastyrskyi A, Roush WR, et al. Therapeutic Targeting of CDK12/CDK13 in Triple-Negative Breast Cancer. Cancer Cell 2019;36(5):545–58 e7 doi 10.1016/j.ccell.2019.09.004. [DOI] [PubMed] [Google Scholar]
- 32.Kohoutek J, Blazek D. Cyclin K goes with Cdk12 and Cdk13. Cell Div 2012;7:12 doi 10.1186/1747-1028-7-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Greifenberg AK, Honig D, Pilarova K, Duster R, Bartholomeeusen K, Bosken CA, et al. Structural and Functional Analysis of the Cdk13/Cyclin K Complex. Cell Rep 2016;14(2):320–31 doi 10.1016/j.celrep.2015.12.025. [DOI] [PubMed] [Google Scholar]
- 34.Bosken CA, Farnung L, Hintermair C, Merzel Schachter M, Vogel-Bachmayr K, Blazek D, et al. The structure and substrate specificity of human Cdk12/Cyclin K. Nat Commun 2014;5:3505 doi 10.1038/ncomms4505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Ekumi KM, Paculova H, Lenasi T, Pospichalova V, Bosken CA, Rybarikova J, et al. Ovarian carcinoma CDK12 mutations misregulate expression of DNA repair genes via deficient formation and function of the Cdk12/CycK complex. Nucleic Acids Res 2015;43(5):2575–89 doi 10.1093/nar/gkv101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Wu YM, Cieslik M, Lonigro RJ, Vats P, Reimers MA, Cao X, et al. Inactivation of CDK12 Delineates a Distinct Immunogenic Class of Advanced Prostate Cancer. Cell 2018;173(7):1770–82 e14 doi 10.1016/j.cell.2018.04.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Cancer Genome Atlas Research N. Integrated genomic analyses of ovarian carcinoma. Nature 2011;474(7353):609–15 doi 10.1038/nature10166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med 2011;364(26):2507–16 doi 10.1056/NEJMoa1103782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Hyman DM, Puzanov I, Subbiah V, Faris JE, Chau I, Blay JY, et al. Vemurafenib in Multiple Nonmelanoma Cancers with BRAF V600 Mutations. N Engl J Med 2015;373(8):726–36 doi 10.1056/NEJMoa1502309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Kopetz S, Desai J, Chan E, Hecht JR, O’Dwyer PJ, Maru D, et al. Phase II Pilot Study of Vemurafenib in Patients With Metastatic BRAF-Mutated Colorectal Cancer. J Clin Oncol 2015;33(34):4032–8 doi 10.1200/JCO.2015.63.2497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Chila R, Chiappa M, Guffanti F, Panini N, Conconi D, Rinaldi A, et al. Stable CDK12 Knock-Out Ovarian Cancer Cells Do Not Show Increased Sensitivity to Cisplatin and PARP Inhibitor Treatment. Front Oncol 2022;12:903536 doi 10.3389/fonc.2022.903536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Sokol ES, Pavlick D, Frampton GM, Ross JS, Miller VA, Ali SM, et al. Pan-Cancer Analysis of CDK12 Loss-of-Function Alterations and Their Association with the Focal Tandem-Duplicator Phenotype. Oncologist 2019;24(12):1526–33 doi 10.1634/theoncologist.2019-0214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Jonsson P, Bandlamudi C, Cheng ML, Srinivasan P, Chavan SS, Friedman ND, et al. Tumour lineage shapes BRCA-mediated phenotypes. Nature 2019;571(7766):576–9 doi 10.1038/s41586-019-1382-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Sun R, Wei T, Ding D, Zhang J, Chen S, He HH, et al. CYCLIN K down-regulation induces androgen receptor gene intronic polyadenylation, variant expression and PARP inhibitor vulnerability in castration-resistant prostate cancer. Proc Natl Acad Sci U S A 2022;119(39):e2205509119 doi 10.1073/pnas.2205509119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Johnson SF, Cruz C, Greifenberg AK, Dust S, Stover DG, Chi D, et al. CDK12 Inhibition Reverses De Novo and Acquired PARP Inhibitor Resistance in BRCA Wild-Type and Mutated Models of Triple-Negative Breast Cancer. Cell Rep 2016;17(9):2367–81 doi 10.1016/j.celrep.2016.10.077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Shyamsunder P, Sridharan SP, Madan V, Dakle P, Zeya C, Kanojia D, et al. THZ531 Induces a State of BRCAness in Multiple Myeloma Cells: Synthetic Lethality with Combination Treatment of THZ 531 with DNA Repair Inhibitors. Int J Mol Sci 2022;23(3) doi 10.3390/ijms23031207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Niu T, Li K, Jiang L, Zhou Z, Hong J, Chen X, et al. Noncovalent CDK12/13 dual inhibitors-based PROTACs degrade CDK12-Cyclin K complex and induce synthetic lethality with PARP inhibitor. Eur J Med Chem 2022;228:114012 doi 10.1016/j.ejmech.2021.114012. [DOI] [PubMed] [Google Scholar]
- 48.Orhan E, Velazquez C, Tabet I, Fenou L, Rodier G, Orsetti B, et al. CDK inhibition results in pharmacologic BRCAness increasing sensitivity to olaparib in BRCA1-WT and olaparib resistant in Triple Negative Breast Cancer. Cancer Lett 2024;589:216820 doi 10.1016/j.canlet.2024.216820. [DOI] [PubMed] [Google Scholar]
- 49.Yang Y, Badura ML, O’Leary PC, Delavan HM, Robinson TM, Egusa EA, et al. Large tandem duplications in cancer result from transcription and DNA replication collisions. medRxiv 2024. doi 10.1101/2023.05.17.23290140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Petropoulos M, Karamichali A, Rossetti GG, Freudenmann A, Iacovino LG, Dionellis VS, et al. Transcription-replication conflicts underlie sensitivity to PARP inhibitors. Nature 2024;628(8007):433–41 doi 10.1038/s41586-024-07217-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Fallah J, Xu J, Weinstock C, Gao X, Heiss BL, Maguire WF, et al. Efficacy of Poly(ADP-ribose) Polymerase Inhibitors by Individual Genes in Homologous Recombination Repair Gene-Mutated Metastatic Castration-Resistant Prostate Cancer: A US Food and Drug Administration Pooled Analysis. J Clin Oncol 2024;42(14):1687–98 doi 10.1200/JCO.23.02105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Clarke NW, Armstrong AJ, Thiery-Vuillemin A, Oya M, Shore N, Loredo E, et al. Abiraterone and Olaparib for Metastatic Castration-Resistant Prostate Cancer. NEJM Evid 2022;1(9):EVIDoa2200043 doi 10.1056/EVIDoa2200043. [DOI] [PubMed] [Google Scholar]
- 53.de Bono J, Mateo J, Fizazi K, Saad F, Shore N, Sandhu S, et al. Olaparib for Metastatic Castration-Resistant Prostate Cancer. N Engl J Med 2020;382(22):2091–102 doi 10.1056/NEJMoa1911440. [DOI] [PubMed] [Google Scholar]
- 54.Hussain M, Mateo J, Fizazi K, Saad F, Shore N, Sandhu S, et al. Survival with Olaparib in Metastatic Castration-Resistant Prostate Cancer. N Engl J Med 2020;383(24):2345–57 doi 10.1056/NEJMoa2022485. [DOI] [PubMed] [Google Scholar]
- 55.de Bono JS, Mehra N, Scagliotti GV, Castro E, Dorff T, Stirling A, et al. Talazoparib monotherapy in metastatic castration-resistant prostate cancer with DNA repair alterations (TALAPRO-1): an open-label, phase 2 trial. Lancet Oncol 2021;22(9):1250–64 doi 10.1016/S1470-2045(21)00376-4. [DOI] [PubMed] [Google Scholar]
- 56.Chi KN, Rathkopf D, Smith MR, Efstathiou E, Attard G, Olmos D, et al. Niraparib and Abiraterone Acetate for Metastatic Castration-Resistant Prostate Cancer. J Clin Oncol 2023;41(18):3339–51 doi 10.1200/JCO.22.01649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Abida W, Campbell D, Patnaik A, Bryce AH, Shapiro J, Bambury RM, et al. Rucaparib for the Treatment of Metastatic Castration-resistant Prostate Cancer Associated with a DNA Damage Repair Gene Alteration: Final Results from the Phase 2 TRITON2 Study. Eur Urol 2023;84(3):321–30 doi 10.1016/j.eururo.2023.05.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Chi KN, Sandhu S, Smith MR, Attard G, Saad M, Olmos D, et al. Niraparib plus abiraterone acetate with prednisone in patients with metastatic castration-resistant prostate cancer and homologous recombination repair gene alterations: second interim analysis of the randomized phase III MAGNITUDE trial. Ann Oncol 2023;34(9):772–82 doi 10.1016/j.annonc.2023.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Fizazi K, Azad AA, Matsubara N, Carles J, Fay AP, De Giorgi U, et al. First-line talazoparib with enzalutamide in HRR-deficient metastatic castration-resistant prostate cancer: the phase 3 TALAPRO-2 trial. Nat Med 2023. doi 10.1038/s41591-023-02704-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Antonarakis ES, Isaacsson Velho P, Fu W, Wang H, Agarwal N, Sacristan Santos V, et al. CDK12-Altered Prostate Cancer: Clinical Features and Therapeutic Outcomes to Standard Systemic Therapies, Poly (ADP-Ribose) Polymerase Inhibitors, and PD-1 Inhibitors. JCO Precis Oncol 2020;4:370–81 doi 10.1200/po.19.00399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Popova T, Manie E, Boeva V, Battistella A, Goundiam O, Smith NK, et al. Ovarian Cancers Harboring Inactivating Mutations in CDK12 Display a Distinct Genomic Instability Pattern Characterized by Large Tandem Duplications. Cancer Res 2016;76(7):1882–91 doi 10.1158/0008-5472.CAN-15-2128. [DOI] [PubMed] [Google Scholar]
- 62.Tien JC, Chang Y, Zhang Y, Chou J, Cheng Y, Wang X, et al. CDK12 Loss Promotes Prostate Cancer Development While Exposing Vulnerabilities to Paralog-Based Synthetic Lethality. bioRxiv 2024. doi 10.1101/2024.03.20.585990. [DOI] [Google Scholar]
- 63.Rescigno P, Gurel B, Pereira R, Crespo M, Rekowski J, Rediti M, et al. Characterizing CDK12-Mutated Prostate Cancers. Clin Cancer Res 2021;27(2):566–74 doi 10.1158/1078-0432.CCR-20-2371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Nguyen CB, Reimers MA, Perera C, Abida W, Chou J, Feng FY, et al. Evaluating Immune Checkpoint Blockade in Metastatic Castration-Resistant Prostate Cancers with Deleterious CDK12 Alterations in the Phase 2 IMPACT Trial. Clin Cancer Res 2024. doi 10.1158/1078-0432.CCR-24-0400. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The whole exome sequencing (WES) data generated in this study are publicly available in the NIH Sequence Read Archive (SRA) under BioProject ID numbers PRJNA1145981 and PRJNA932332. Data from the TRITON2 clinical trial are available upon request from the corresponding authors. Drug screening data and western blots can be found within the article and its supplementary data files, and raw data for plots are available upon request from the corresponding authors. Plasmids and cell lines described in this study are available upon request from the corresponding authors.