search for   

 

Synergistic Effects of Combined PROTAC-based EZH2 Degrader and METTL3 Inhibitor in Burkitt’s Lymphoma
Korean J Clin Lab Sci 2024;56:198-206  
Published on September 30, 2024
Copyright © 2024 Korean Society for Clinical Laboratory Science.

Minseo YU , Ra Eun KIM , Yurim JEONG , Hyewon JANG , Se Been KIM , Jung-Yeon LIM

Department of Biomedical Laboratory Science, Inje University, Gimhae, Korea
Correspondence to: Jung-Yeon LIM
Department of Biomedical Laboratory Science, Inje University, 197 Inje-ro, Gimhae 50834, Korea
E-mail: limjy@inje.ac.kr
ORCID: https://orcid.org/0000-0001-5903-8810
This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
Abstract
EZH2 is a methyltransferase that is a critical target for lymphoma treatment. However, it is not yet widely used in clinical settings. PROteolysis TArgeting Chimeras (PROTACs) represent a novel therapeutic strategy aimed at eliminating proteins that have been a challenging target using conventional small molecules. In our previous research, we compared the small molecules-based EZH2 inhibitor used in clinical settings with a PROTAC-based EZH2 degrader. We found that the PROTAC-based degrader was significantly more effective. Building on this, we further investigated the effects of combining the PROTAC-based EZH2 degrader (dEZH2) with a METTL3 inhibitor, both of which have demonstrated effectiveness in inhibiting cell proliferation and inducing apoptosis in Burkitt’s lymphoma. Using the CCK-8 assay, we found that both drugs, alone and in combination, significantly inhibited Daudi and Ramos cell growth in a dose-dependent manner. The combined treatment markedly suppressed cell proliferation and induced apoptosis, as confirmed by Annexin V/PI staining. Our results revealed G2/M phase arrest with a significant decrease in the G0/G1 phase by flow cytometry. Our study also showed increased levels of cleaved PARP, cleaved caspase-3, tumor protein p53 (TP53), and PUMA using the western blot technique, indicating enhanced p53-dependent apoptosis. Our findings suggest that the combination therapy of dEZH2 and iMETTL3 could be a promising approach in the treatment of Burkitt’s lymphoma.
Keywords : Apoptosis, Burkitt’s lymphoma, Enhancer of zeste homolog 2 protein, METTL3 protein, Proteolysis targeting chimera
INTRODUCTION

Small molecule drug discovery has primarily focused on directly regulating protein activity, especially through inhibitors. Recently, targeted protein degradation (TPD) technologies have emerged utilizing the cell’s own degradation mechanisms using PROteolysis TArgeting Chimeras (PROTACs) [1], molecular glues, Lysosome-Targeting Chimeras [2], and Antibody-based PROTACs [3] to eliminate disease-associated proteins. These advances have not only broadened the scope of new target drug discovery but also enabled the pharmaceutical industry to tackle previously ‘undruggable’ targets, with significant clinical progress demonstrated in cancer therapies [4, 5].

Based on our previous research, we showed to demonstrate and compare the inhibitory effects of two drugs: the enhancer of zeste homolog 2 (EZH2) inhibitor Tazemetostat (iEZH2) and the novel PROTAC-based degrader MS1943 (dEZH2) [6]. Our results demonstrated that dEZH2 was significantly more effective at suppressing B cell lymphoma than the iEZH2 [7, 8]. Also, combination of dEZH2 with the Ibrutinib (Bruton’s tyrosine kinase inhibitor) led to a synergistic reduction in B cell lymphoma cells [7]. Additionally, dEZH2 showed effectiveness in inhibiting Burkitt’s lymphoma when used in combination with Lapatinib, an epidermal growth factor receptor/human epidermal growth factor receptor 2 targeted therapy [9]. These findings show that PROTAC-based drugs have significant therapeutic potential for targeting the same protein, offering promising new treatment strategies.

EZH2 is a methyltransferase, which contributes to tumor progression by silencing tumor suppressor genes, while methyltransferase 3 (METTL3) regulates mRNA modifications that promote oncogenic gene expression. Previous reports suggested that METTL3 modulated protein levels of EZH2 through m6A modification [10, 11]. By targeting both, we could more effectively disrupt the pathway that drive Burkitt’s lymphoma cell proliferation and potentially overcome resistance to single-drug therapy. METTL3 inhibitors provides the potential therapeutic targeting cancer therapy. The anticancer effects of METTL3 inhibitors have been demonstrated in diffuse large B-cell lymphoma [12, 13], acute myeloid leukemia (AML) [14, 15] and many different types of blood type cancer.

Burkitt’s lymphoma is a highly aggressive type of lymphoma that targets B-lymphocytes and constitutes about less 1% of all non-Hodgkin lymphomas [16, 17]. This study aims to demonstrate the efficacy of combining the PROTAC-based EZH2 degrader with a METTL3 inhibitor (iMETTL3) in a Burkitt’s lymphoma cell lines.

MATERIALS AND METHODS

1. Cell Lines and Cell Culture

In this study, Burkitt’s lymphoma cell lines including, Daudi and Ramos cells were used. These cell lines were obtained from Korean Cell Line Bank. These cell lines were grown in RPMI 1640 medium (Gibco) supplemented with 10% heat-inactivated fetal bovine serum (FBS; Gibco), 2mM L-glutamine (Gibco), 1% antibiotics (10 U/mL penicillin and 10 g/mL streptomycin; Gibco). All cell lines were incubated at 37℃ and 5% CO2.

2. Drug Preparations

EZH2 degrader (MS1943) and METTL3 inhibitor (STM2457) were purchased from MedChemExpress. Both were dissolved in dimethyl sulfoxide (DMSO) as recommended by the manufacturer and stored at –80℃. All these drugs were diluted with the RPMI 1640 medium with 10% heat-inactivated FBS, 2 mM L-glutamine, and 1% antibiotics before being used for the treatment of cell lines.

3. Cell Proliferation Assay

Cell growth was assessed using the Cell Counting Kit-8 (CCK-8; Dojindo) assay according to the manufacturer’s protocol. Daudi and Ramos cell lines were seeded at an initial cell density of 2×104 cells/100 μL culture medium in 96-well plates. These cells were treated with various doses of EZH2 degrader and METTL3 inhibitor drugs without any drugs, 2.5 μM, 5 μM, and 10 μM for each drug. The synergistic effects were assessed using 5 μM for both drugs. Cultures were maintained at 37℃ in a 5% CO2 atmosphere. CCK-8 solution was added in increments of 10 μL to each well after 24 hours, 48 hours, and 72 hours. The plates were incubated for 1∼4 hours in a CO2 incubator and the optical density was measured at 450 nm using a microplate reader.

4. Western Blotting

One million Ramos or Daudi cells were lysed in 2× laemmli sample buffer (Bio-Rad) with β-mercaptoethanol and boiled at 95℃ for 10 minutes. After removal of the insoluble fraction by centrifugation at 10,000×g for 10 min, protein samples were separated by SDS gel electrophoresis and transferred to a polyvinylidene difluoride membrane. The membranes were stained with cleaved poly (ADP-ribose) polymerase (PARP), PARP, cleaved caspase-3, tumor protein P53 (TP53) and p53 upregulated modulator of apoptosis (PUMA) antibodies (Cell Signaling Technology) at a dilution of 1:1,000 or GAPDH antibody (Cell Signaling Technology) at a dilution of 1:5,000 at 4℃ overnight. After overnight incubation at 4℃, HRP-conjugated secondary antibodies were added. After washing with Tris-buffered saline and Tween 20, the hybridized bands were detected using an enhanced chemiluminescence (ECL) detection kit (Amersham Pharmacia Biotech).

5. Cell Cycle Analysis

The 5×105 cells were treated with the drugs described. After incubating for 72 hours in CO2 incubator, the cells were harvested and centrifuged 2,000 rpm 5 minutes with DPBS (Gibco). These cells were performed twice for wash and discarded the supernatant. For cell cycle analysis, the Annexin V-propidium iodide (PI) Apoptosis Kit (BioVision) was used following the manufacturer’s protocol. The cell cycle was detected using the Novocyte Advanteon (Agilent).

6. Statistical Analysis

All data were statistically evaluated by Student’s t-test using GraphPad Prism 8 (GraphPad Software Inc.). All experiments were conducted at least thrice. The values are depicted using the mean and SEM. Differences between the means of the groups were considered to be statistically significant if P<0.05, P<0.01, and P<0.001.

RESULTS

1. dEZH2 and iMETTL3 Suppress Cell Growth in Burkitt’s Lymphoma Cell Lines in a Dose-dependent Manner at 24, 48, and 72 Hours Post Treatment

We examined the effects of dEZH2 and iMETTL3 on the proliferation of Daudi and Ramos cells using the CCK-8 assay. Both cell lines were treated with various concentrations of the EZH2 degrader and METTL3 inhibitor (ranging from 2.5 to 10 μM) at different time points: 24 hours (Figure 1A), 48 hours (Figure 1B), and 72 hours (Figure 1C). And we showed dEZH2, iMETTL3 and comb cell viability (%) in same concentration by time period in Daudi (Figure 1D) and Ramos (Figure 1E). Our findings clearly demonstrated that these treatments significantly inhibited the proliferation of Burkitt’s lymphoma cells.

Fig. 1. Dose-dependent cell survival rate of dEZH2 and iMETTL3 in Burkitt’s lymphoma. Daudi and Ramos cells were treated with 2.5, 5 and 10 μM of dEZH2 or iMETTL3 for (A) 24, (B) 48, and (C) 72 hours, with dimethyl sulfoxide as a control. And, dEZH2 5 μM, iMETTL3 5 μM and combination (dEZH2 5 μM+iMETTL3 5 μM) treatments were compared cell viability (%) by time points in Daudi (D) and Ramos (E). Statistical testing was conducted with two-tailed, unpaired t-tests: *P<0.05; **P<0.01; ***P<0.001. Error bars represent the ±SD.
Abbreviations: dEZH2, PROteolysis TArgeting Chimeras-based enhancer of zeste homolog 2 degrader; iMETTL3, methyltransferase-like 3 inhibitor.

2. Combination of dEZH2 and iMETTL3 Significantly Suppresses Cell Proliferation in Burkitt’s Lymphoma Cells

To evaluate the inhibitory effects of dEZH2 and iMETTL3 on the proliferation of Burkitt’s lymphoma cells in vitro, we performed cell proliferation assays on cultured Daudi and Ramos cells, with or without these drugs. Our results demonstrated that the combination of the dEZH2 and iMETTL3 significantly reduced cell growth compared to individual treatments at the 24 hours (Figure 2A), 48 hours (Figure 2B), and 72 hours (Figure 2C). We also performed the toxicity assays in HEK293T cells showed no significant difference with dEZH2 and iMETTL3 (data not shown). These findings suggest that the combined therapy effectively suppresses the survival and proliferation of Burkitt’s lymphoma cells, indicating a synergistic effect.

Fig. 2. Combination treatments of dEZH2 with or iMETTL3 in Burkitt’s lymphoma. Cell viability of Daudi and Ramos cell lines was analyzed using Cell Counting Kit-8. Both cell lines were plated at 1×104/well and treated with 5 μM of each drug, or a combination of dEZH2 and iMETTL3 for (A) 24, (B) 48, and (C) 72 hours, with dimethyl sulfoxide as a control. Statistical testing was conducted with two-tailed, unpaired t-tests: *P<0.05; **P<0.01; ***P<0.001. Error bars represent the ±SD.
Abbreviations: dEZH2, PROteolysis TArgeting Chimeras-based enhancer of zeste homolog 2 degrader; iMETTL3, methyltransferase-like 3 inhibitor.

3. Combination of dEZH2 and iMETTL3 Treatment Significantly Enhances Apoptosis in Burkitt’s Lymphoma Cells

To verify that dEZH2 degrader and iMETTL3 induce cell death, we conducted annexin V/PI staining analysis. The data indicated a pronounced combined effect of the drugs in the Q2 (late apoptosis) and Q3 (early apoptosis) quadrants. The combination treatment promoted both early and late apoptosis in Daudi (Figure 3A, 3B) and Ramos cells (Figure 3C, 3D).

Fig. 3. A synergistic effect on apoptosis was observed in combination therapy for Burkitt’s lymphoma. Annexin V/PI staining was used to determine the apoptosis rate of (A, B) Daudi cell and (C, D) Ramos cell lines. The control group was exposed to DMSO, while the rest were treated with 5 μM of dEZH2 and iMETTL3 individually or in combination for 72 hours. (B, D) Viable cells (Annexin V-negative/PI-negative), early apoptotic cells (Annexin V-positive/PI-negative), and late apoptotic cells (Annexin V-positive/PI-positive) were measured using flow cytometry. Statistical testing was conducted with two-tailed, unpaired t-tests: *P<0.05; **P<0.01; ***P<0.001. Error bars represent the ±SD.
Abbreviations: DMSO, dimethyl sulfoxide; dEZH2, PROteolysis TArgeting Chimeras-based enhancer of zeste homolog 2 degrader; iMETTL3, methyltransferase-like 3 inhibitor; PI, propidium iodide.

4. Combination of dEZH2 and iMETTL3 Treatment Induces G2/M-phase Arrest in Burkitt’s Lymphoma Cells

To investigate the relationship between cell proliferation inhibition and cell cycle regulation, we performed flow cytometric analysis to assess cell distribution across different cell cycle phases. Treatment with a combination of dEZH2 and iMETTL3 resulted in a significant increase in Burkitt’s lymphoma cells in the G2/M phase after 72 hours. Concurrently, there was a decrease in the number of cells in the G0/G1 phase in both Daudi (Figure 4A) and Ramos cells (Figure 4B).

Fig. 4. Combination therapy with dEZH2 and iMETTL3 results in increased S and G2∼M phase arrest in flow cytometric analysis of cell cycle phases. The percentage of cell cycle stages was determined using flow cytometry. (A) In each cell cycle phase, the main peaks of the Daudi cell line are shown from left to right: G0/G1 phase, S phase, and G2/M phase. (B) Ramos cell line was exposed to 5 μM of dEZH2 and iMETTL3 for 72 hours. Cell cycle stages were analyzed as percentage ratios, excluding some cells outside the measurement range. Statistical testing was conducted with two-tailed, unpaired t-tests. Error bars represent the ±SD.
Abbreviations: DMSO, dimethyl sulfoxide; dEZH2, PROteolysis TArgeting Chimeras-based enhancer of zeste homolog 2 degrader; iMETTL3, methyltransferase-like 3 inhibitor.

5. Combination Treatment of dEZH2 and iMETTL3 Causes Apoptosis through p53-dependent Pathway

After confirming apoptosis induction with the dual inhibition of EZH2 and METTL3, we examined the involvement of specific apoptosis-related pathways using western blot analysis. PARP, a key protein in DNA repair, becomes cleaved upon activation by Caspase-3, leading to DNA damage and apoptosis [18, 19]. Compared to the control group, Daudi cells showed significantly increased levels of cleaved PARP and cleaved caspase-3 (Figure 5A). We further investigated the role of the pro-apoptotic gene TP53, which induces cell cycle arrest and activates PUMA, promoting cell death in cancer cells [20, 21]. The combination therapy increasd the expression of TP53 and PUMA in Daudi cells (Figure 5B). In summary, our study demonstrates that the combination therapy enhances p53-dependent apoptosis, as indicated by the upregulation of cleaved PARP, cleaved caspase-3, TP53, and PUMA.

Fig. 5. Apoptosis-related protein levels increased in Daudi cell treated with a combination of dEZH2 and iMETTL3. The western blot method was used to examine specific protein expression levels in Daudi cell lines treated with 5 μM of dEZH2 and iMETTL3, either individually or in combination. (A) Protein expression levels of c-Caspase-3, PARP, and c-PARP in Daudi cell line were analyzed compared with GAPDH. (B) Protein expression levels of TP53 and TP53-related PUMA were analyzed relative to GAPDH. Statistical testing was conducted with two-tailed, unpaired t-tests. Error bars represent the ±SD.
Abbreviations: dEZH2, PROteolysis TArgeting Chimeras-based enhancer of zeste homolog 2 degrader; iMETTL3, methyltransferase-like 3 inhibitor; c-Caspase-3, cleaved caspase-3; PARP, poly (ADP-ribose) polymerase; c-PARP, cleaved PARP; GAPDH, glyceraldehyde 3-phosphate dehydrogenase; PUMA, p53 upregulated modulator of apoptosis; TP53, tumor protein P53.
DISCUSSION

PROTAC-based drugs offer several distinct advantages over traditional small molecule inhibitors. First, PROTAC-based drugs induce TPD, leading to the complete removal of disease-causing proteins, which ensures a sustained therapeutic effect since the target protein needs to be resynthesized to regain functionality [5, 22]. Second, this approach can overcome drug resistance, as PROTAC-based drugs can degrade mutated proteins that are not effectively inhibited by conventional small molecules [23]. Third, PROTAC-based drugs can target a broader range of protein conformations and mutations, enhancing their therapeutic versatility. Additionally, PROTACs often require lower dosages than traditional inhibitors, potentially reducing side effects [24]. These advantages collectively underscore the transformative potential of PROTACs in developing more effective and durable cancer treatments [25, 26].

Our previous research demonstrated the superior efficacy of the PROTAC-based EZH2 degrader, MS1943, compared to the clinical EZH2 inhibitor, Tazemetostat in B cell lymphoma cells [27, 28]. In this study, we extended these findings by exploring the synergistic effects of combining EZH2 degrader with the METTL3 inhibitor on Burkitt’s lymphoma cells. The combination therapy was strikingly reduced in Ramos and Daudi cell proliferation and significantly increased levels of apoptosis related protein, highlighting the therapeutic potential of PROTAC-based strategies.

The results of our study provide compelling evidence that the combination of dEZH2 and iMETTL3 can effectively suppress the growth of Burkitt’s lymphoma cells and induce apoptosis through a p53-dependent pathway. This results are significant given the challenges associated with targeting EZH2 in clinical settings. And, we showed that treatment with a combination of dEZH2 and iMETTL3 resulted in a significant increase in the G2/M phase. We also observed an increase in the S phase of the cell cycle following combination treatment. These results suggest that inhibition of METTL3 increases PLK1 expression, leading to accumulation in the S phase and potentially causing arrest in the G2/M phase.

The clinical implications of these findings are important in Burkitt’s lymphoma therapy. The enhanced efficacy of the combination of dEZH2 and iMETTL3 offers a promising new approach for Burkitt’s lymphoma therapy. The ability of this combination to target and degrade disease-associated proteins more effectively than conventional inhibitors underscores the potential of TPD technologies in oncology.

Moreover, the successful use of this combination therapy need to perform the preclinical models for potential clinical trials. The broader application of PROTAC-based strategies need in various cancer types such as breast cancer, lung cancer, and prostate where EZH2 is overexpressed, particularly those that have been challenging to treat with traditional small molecule inhibitor [29, 30].

In conclusion, our study demonstrates that the combination dEZH2 degrader and iMETTL3 significantly suppresses Burkitt’s lymphoma cell growth and induces p53-dependent apoptosis pathway. These findings show a strong rationale for the continued development and clinical evaluation of PROTAC-based therapies, providing new hope for improved treatment outcomes in Burkitt’s lymphoma patients and potentially other hematologic malignancy.

요 약

PROteolysis TArgeting Chimeras (PROTACs)은 기존의 저분자(small moleule)로는 타겟팅이 어려웠던 단백질을 제거하기 위한 새로운 치료 전략을 제공한다. EZH2는 림프종 치료의 중요한 타겟인 메틸트랜스퍼라제(methyltransferase)이지만, 임상에서 널리 사용되지 않는다. 이전 연구에서 우리는 임상에서 사용되는 EZH2 억제제와 PROTAC 기반의 EZH2 분해제를 비교했으며, PROTAC 기반 분해제가 훨씬 더 효과적임을 확인했다. 이를 바탕으로, 우리는 PROTAC 기반의 EZH2 분해제와 기존 림프종 치료제인 METTL3 억제제를 병용하여 버킷림프종 세포의 증식과 세포 사멸에 미치는 영향을 조사했다. CCK-8 분석을 통해 두 약물이 단독 및 병용 처리 시 농도 의존적으로 Daudi 및 Ramos 세포 성장을 유의미하게 억제하는 것을 확인했다. 병용 치료는 세포 증식을 현저히 억제하고, annexin V/PI 염색을 통해 세포 사멸을 유도하는 것으로 확인되었다. 유세포 분석에서는 G0/G1 단계의 감소와 함께 G2/M 단계에서의 세포 주기 정지를 보였다. 웨스턴 블럿(western blot) 분석에서는 cleaved PARP, cleaved caspase-3, TP53 및 PUMA의 증가된 수준을 나타내어 p53 의존적인 세포 사멸이 강화됨을 증명했다. 우리는 이 연구 결과가 이 병용 요법이 버킷림프종 치료에 유망한 접근법임을 제안한다.

Acknowledgements

None

Funding

This work was supported by grant from Inje University, 2023 (No. 20230017).

Conflict of interest

None

Author’s information (Position)

Yu M, Graduate student; Kim RE, Graduate student; Jeong Y, Master graduate student; Jang H, Graduate student; Kim SB, Graduate student; Lim JY, Professor.

Author Contributions

-Conceptualization: Yu M, Kim RE, Jeong Y.

-Data curation: Yu M, Kim RE, Jeong Y.

-Formal analysis: Yu M, Kim RE.

-Methodology: Jeong Y, Kim SB.

-Validation: Lim JY.

-Writing - original draft: Lim JY, Yu M, Kim RE.

-Writing - review & editing: Yu M, Kim RE, Jang H, Jeong Y, Lim JY.

Ethics approval

This article does not require IRB/IACUC approval because there are no human and animal participants.

References
  1. Chen S, Cui J, Chen H, Yu B, Long S. Recent progress in degradation of membrane proteins by PROTACs and alternative targeted protein degradation techniques. Eur J Med Chem. 2023;262:115911. https://doi.org/10.1016/j.ejmech.2023.115911
    Pubmed CrossRef
  2. Paudel RR, Lu D, Roy Chowdhury S, Monroy EY, Wang J. Targeted protein degradation via lysosomes. Biochemistry. 2023;62:564-579. https://doi.org/10.1021/acs.biochem.2c00310
    Pubmed KoreaMed CrossRef
  3. Crunkhorn S. Developing antibody-based PROTACs. Nat Rev Drug Discov. 2022;21:795. https://doi.org/10.1038/d41573-022-00159-2
    Pubmed CrossRef
  4. Békés M, Langley DR, Crews CM. PROTAC targeted protein degraders: the past is prologue. Nat Rev Drug Discov. 2022;21:181-200. https://doi.org/10.1038/s41573-021-00371-6
    Pubmed KoreaMed CrossRef
  5. Li X, Song Y. Proteolysis-targeting chimera (PROTAC) for targeted protein degradation and cancer therapy. J Hematol Oncol. 2020;13:50. https://doi.org/10.1186/s13045-020-00885-3
    Pubmed KoreaMed CrossRef
  6. Ma A, Stratikopoulos E, Park KS, Wei J, Martin TC, Yang X, et al. Discovery of a first-in-class EZH2 selective degrader. Nat Chem Biol. 2020;16:214-222. https://doi.org/10.1038/s41589-019-0421-4
    Pubmed KoreaMed CrossRef
  7. Jeong Y, Kim SB, Yang CE, Yu MS, Choi WS, Jeon Y, et al. Overcoming the therapeutic limitations of EZH2 inhibitors in Burkitt's lymphoma: a comprehensive study on the combined effects of MS1943 and Ibrutinib. Front Oncol. 2023;13:1252658. https://doi.org/10.3389/fonc.2023.1252658
    CrossRef
  8. Xie H, Xu W, Liang J, Liu Y, Zhuo C, Zou X, et al. Design, synthesis and evaluation of EZH2-based PROTACs targeting PRC2 complex in lymphoma. Bioorg Chem. 2023;140:106762. https://doi.org/10.1016/j.bioorg.2023.106762
    Pubmed CrossRef
  9. Kim SB, Yang CE, Jeong Y, Yu M, Choi WS, Lim JY, et al. Dual targeting of EZH2 degradation and EGFR/HER2 inhibition for enhanced efficacy against Burkitt's lymphoma. Cancers (Basel). 2023;15:4472. https://doi.org/10.3390/cancers15184472
    Pubmed KoreaMed CrossRef
  10. Zeng C, Huang W, Li Y, Weng H. Roles of METTL3 in cancer: mechanisms and therapeutic targeting. J Hematol Oncol. 2020;13:117. https://doi.org/10.1186/s13045-020-00951-w
    Pubmed KoreaMed CrossRef
  11. Li Y, He X, Lu X, Gong Z, Li Q, Zhang L, et al. METTL3 acetylation impedes cancer metastasis via fine-tuning its nuclear and cytosolic functions. Nat Commun. 2022;13:6350. https://doi.org/10.1038/s41467-022-34209-5
    Pubmed KoreaMed CrossRef
  12. Li J, Zhu Z, Zhu Y, Li J, Li K, Zhong W. METTL3-mediated m6A methylation of C1qA regulates the Rituximab resistance of diffuse large B-cell lymphoma cells. Cell Death Discov. 2023;9:405. https://doi.org/10.1038/s41420-023-01698-2
    Pubmed KoreaMed CrossRef
  13. Meng S, Xia Y, Li M, Wu Y, Wang D, Zhou Y, et al. NCBP1 enhanced proliferation of DLBCL cells via METTL3-mediated m6A modification of c-Myc. Sci Rep. 2023;13:8606. https://doi.org/10.1038/s41598-023-35777-2
    Pubmed KoreaMed CrossRef
  14. Li M, Ye J, Xia Y, Li M, Li G, Hu X, et al. METTL3 mediates chemoresistance by enhancing AML homing and engraftment via ITGA4. Leukemia. 2022;36:2586-2595. https://doi.org/10.1038/s41375-022-01696-w
    Pubmed KoreaMed CrossRef
  15. Yankova E, Blackaby W, Albertella M, Rak J, De Braekeleer E, Tsagkogeorga G, et al. Small-molecule inhibition of METTL3 as a strategy against myeloid leukaemia. Nature. 2021;593:597-601. https://doi.org/10.1038/s41586-021-03536-w
    Pubmed KoreaMed CrossRef
  16. Ferry JA. Burkitt's lymphoma: clinicopathologic features and differential diagnosis. Oncologist. 2006;11:375-383. https://doi.org/10.1634/theoncologist.11-4-375
    CrossRef
  17. Schmitz R, Ceribelli M, Pittaluga S, Wright G, Staudt LM. Oncogenic mechanisms in Burkitt lymphoma. Cold Spring Harb Perspect Med. 2014;4:a014282. https://doi.org/10.1101/cshperspect.a014282
    Pubmed CrossRef
  18. Los M, Mozoluk M, Ferrari D, Stepczynska A, Stroh C, Renz A, et al. Activation and caspase-mediated inhibition of PARP: a molecular switch between fibroblast necrosis and apoptosis in death receptor signaling. Mol Biol Cell. 2002;13:978-988. https://doi.org/10.1091/mbc.01-05-0272
    Pubmed KoreaMed CrossRef
  19. Boulares AH, Yakovlev AG, Ivanova V, Stoica BA, Wang G, Iyer S, et al. Role of poly(ADP-ribose) polymerase (PARP) cleavage in apoptosis. Caspase 3-resistant PARP mutant increases rates of apoptosis in transfected cells. J Biol Chem. 1999;274:22932-22940. https://doi.org/10.1074/jbc.274.33.22932
    Pubmed CrossRef
  20. Yu J, Zhang L. PUMA, a potent killer with or without p53. Oncogene. 2008;27 Suppl 1(Suppl 1):S71-S83. https://doi.org/10.1038/onc.2009.45
    CrossRef
  21. Han CW, Lee HN, Jeong MS, Park SY, Jang SB. Structural basis of the p53 DNA binding domain and PUMA complex. Biochem Biophys Res Commun. 2021;548:39-46. https://doi.org/10.1016/j.bbrc.2021.02.049
    Pubmed CrossRef
  22. Qi SM, Dong J, Xu ZY, Cheng XD, Zhang WD, Qin JJ. PROTAC: an effective targeted protein degradation strategy for cancer therapy. Front Pharmacol. 2021;12:692574. https://doi.org/10.3389/fphar.2021.692574
    Pubmed KoreaMed CrossRef
  23. Neklesa TK, Winkler JD, Crews CM. Targeted protein degradation by PROTACs. Pharmacol Ther. 2017;174:138-144. https://doi.org/10.1016/j.pharmthera.2017.02.027
    Pubmed CrossRef
  24. Liu Z, Hu M, Yang Y, Du C, Zhou H, Liu C, et al. An overview of PROTACs: a promising drug discovery paradigm. Mol Biomed. 2022;3:46. https://doi.org/10.1186/s43556-022-00112-0
    Pubmed KoreaMed CrossRef
  25. Bricelj A, Steinebach C, Kuchta R, Gütschow M, Sosič I. E3 ligase ligands in successful PROTACs: an overview of syntheses and linker attachment points. Front Chem. 2021;9:707317. https://doi.org/10.3389/fchem.2021.707317
    Pubmed KoreaMed CrossRef
  26. Dale B, Cheng M, Park KS, Kaniskan HÜ, Xiong Y, Jin J. Advancing targeted protein degradation for cancer therapy. Nat Rev Cancer. 2021;21:638-654. https://doi.org/10.1038/s41568-021-00365-x
    Pubmed KoreaMed CrossRef
  27. Velez J, Dale B, Park KS, Kaniskan HÜ, Yu X, Jin J. Discovery of a novel, highly potent EZH2 PROTAC degrader for targeting non- canonical oncogenic functions of EZH2. Eur J Med Chem. 2024;267:116154. https://doi.org/10.1016/j.ejmech.2024.116154
    Pubmed KoreaMed CrossRef
  28. Liu Z, Hu X, Wang Q, Wu X, Zhang Q, Wei W, et al. Design and synthesis of EZH2-based PROTACs to degrade the PRC2 complex for targeting the noncatalytic activity of EZH2. J Med Chem. 2021;64:2829-2848. https://doi.org/10.1021/acs.jmedchem.0c02234
    Pubmed CrossRef
  29. Diehl CJ, Ciulli A. Discovery of small molecule ligands for the von Hippel-Lindau (VHL) E3 ligase and their use as inhibitors and PROTAC degraders. Chem Soc Rev. 2022;51:8216-8257. https://doi.org/10.1039/d2cs00387b
    Pubmed KoreaMed CrossRef
  30. An S, Fu L. Small-molecule PROTACs: an emerging and promising approach for the development of targeted therapy drugs. EBioMedicine. 2018;36:553-562. https://doi.org/10.1016/j.ebiom.2018.09.005
    Pubmed KoreaMed CrossRef

Full Text(PDF) Free

Cited By Articles
  • CrossRef (0)

Author ORCID Information

Funding Information