Clinical Efficacy and Tumour Microenvironment Influence of Decitabine Plus R‐CHOP in Patients with Newly Diagnosed Diffuse Large B‐Cell Lymphoma: Phase 1/2 and Biomarker Study
Mu-Chen Zhang,Ying Fang,Peng-Peng Xu,Lei Dong,Rong Shen,Yao-Hui Huang,Di Fu,Zi-Xun Yan,Shu Cheng,Xu-Feng Jiang,Qi Song,Yang He,Yan Zhao,Min Lu,Jing Ye,Feng Liu,Lin Cheng,Chao-Fu Wang,Li Wang,Wei-Li Zhao
DOI: https://doi.org/10.1002/ctm2.584
IF: 8.554
2021-01-01
Clinical and Translational Medicine
Abstract:Epigenetic gene alterations play an important role on diffuse large B cell lymphoma (DLBCL) progression.1 DNA methyltransferase inhibitor (DNMTi) decitabine has demonstrated anti-lymphoma activities, but never been applied for newly diagnosed DLBCL treatment. Here, we conducted for the first time a phase 1/2 trial of decitabine plus standard immunochemotherapy rituximab, cyclophosphamide, doxorubicin, vincristine and prednisone (DR-CHOP, NCT02951728) in DLBCL patients with international prognostic index (IPI) ≥ 2. The study determined the maximum tolerated dose (MTD) of decitabine as 10 mg/m2 on days 1–5 prior to R-CHOP on days 6–11 and showed promising efficacy and good tolerability. The trial enrolled 54 patients, 11 in phase 1 and 43 in phase 2 (Table 1). Among 49 evaluable patients (including six patients received the MTD of decitabine in phase 1), 39 (79.6%) patients achieved complete remission, and six (12.2%) patients achieved partial remission. Two-year progression-free survival (PFS), event-free survival (EFS) and overall survival (OS) rates were 71.4%, 65.3% and 87.8%, respectively (Figure 1A). Intermediate-high (IPI 2–3) or high-risk (IPI 4–5) patients presented similar outcomes (Figure 1B), irrespective on cell of origin and BCL2/MYC double expression (as defined by immunohistochemistry BCL2 ≥ 50% and MYC ≥ 40%) (Figure S1A,B). In our previous cohort of NHL-001 (NCT01852435), 2-year PFS was 59.6%, with OS as 76.2% for IPI ≥ 2 patients with standard R-CHOP (R-CHOP50 and R-CEOP70)2 (Figure 1C). The main adverse events (Table S1) were grade 3–4 hematological toxicity, particularly grade 3–4 neutropenia, comparable to other novel targeted agents plus R-CHOP as ibrutinib, lenalidomide and venetoclax3 and manageable with granulocyte-colony stimulating factor prophylaxis and supportive care. To explore potential biomarkers related to clinical response, DNA-sequencing and RNA-sequencing were performed in patients with quality-controlled tumour samples. Histone/DNA methylation gene mutations occurred in KMT2D (7/46, 15.2%), KMT2C (6/46, 13.0%), TET2 (5/46, 10.9%), HIST1H1C (3/46, 6.5%) and HIST1H1E (3/46, 6.5%). Histone acetylation gene mutations occurred in CREBBP (3/46, 6.5%) and EP300 (2/46, 4.3%). Chromatin remodeling gene mutations occurred in ARID1A (5/46, 10.9%) and SGK1 (2/46, 4.3%). Interferon-γ response pathway gene mutations occurred in SOCS1 (7/46, 15.2%), TP53 (5/46, 10.9%), B2M (4/46, 8.7%), IRF8 (3/46, 6.5%) and CIITA (2/46, 4.3%). T-cell activation gene mutations occurred in PRDM1 (5/46, 10.9%), TNFRSF14 (5/46, 10.9%), BCL6 (4/46, 8.7%), CD70 (3/46, 6.5%) and MPEG1 (2/46, 4.3%). BCR/NF-κB pathway gene mutations occurred in TNFAIP3 (9/46, 19.6%), DTX1 (7/46, 15.2%), MYD88 (7/46, 15.2%), CARD11 (3/46, 6.5%), CD79B (2/46, 4.3%), PTPN6 (2/46, 4.3%) and ZNF608 (2/46, 4.3%). WNT pathway gene mutations occurred in PIM1 (8/46, 17.4%), DDX3X (3/46, 6.5%), GNA13 (3/46, 6.5%) and TBL1XR1 (2/46, 4.3%). JAK-STAT pathway gene mutations occurred in STAT6 (4/46, 8.7%) and STAT3 (2/46, 4.3%). PI3K-AKT pathway gene mutations occurred in ATM (3/46, 6.5%), TSC2 (3/46, 6.5%) and MTOR (2/46, 4.3%). Cell cycle pathway gene mutations occurred in CCND3 (5/46, 10.9%), BTG1 (5/46, 10.9%), BTG2 (4/46, 8.7%), EBF1 (3/46, 6.5%), FAS (2/46, 4.3%) and NFKBIE (2/46, 4.3%) (Figure 2A). Univariate hazard estimates used unadjusted Cox proportional hazards models. Multivariate analysis included clinicopathological parameters and gene mutations demonstrating significance with p < 0.05 on univariate analysis. As expected, adverse prognostic effect of histone/DNA methylation gene mutations was not observed. Interferon-γ response pathway gene mutations were significantly related to prolonged PFS (p = 0.021) and EFS (p = 0.024) (Figure 2B) and independently predicted favorable PFS by multivariate analysis. Among interferon-γ response genes, SOCS1 mutations may induce interferon-γ signaling and increase immune cell activation.4 IRF8 can modulate T-helper cell differentiation and function.5 B2M and CIITA mutations impair human leukocyte antigen-mediated cancer cell recognition and are responsible for cancer immune escape.6 Using RNA-sequencing analysis, gene expression patterns of 14 patients with interferon-γ response pathway gene mutations and 21 patients without mutation were further compared. As confirmed by gene ontology and gene set enrichment analysis (Figures 2C and S2A), multiple signaling pathways were upregulated in the mutation group, including T-helper 1 type immune response, interferon-γ production, response to interferon-γ, T-cell differentiation, T-cell activation and response to tumour necrosis factor pathways. Similar signaling pathway signatures were also observed in 33 DR-CHOP-responding patients (28 complete remission and five partial remission), as compared to four non-responding patients (two stable disease and two progressive disease, Figure S2B,C). This was consistent with previous report that low-dose decitabine (10 mg/day for 5 days) increased circulating interferon-γ-expressing CD3+T cells in Hodgkin's lymphoma.7 Moreover, DNMTi may enhance interferon response in cancer through endogenous retroviruses.8 These findings indicated that the microenvironment influence on interferon-γ response and T-cell activation were closely related to clinical response of DR-CHOP. Functionally, interferon-γ sensitivity of lymphoma cells is enhanced by interferon-γ receptor 2, which is fundamental for anti-tumour response.9 Indeed, as shown in Figure 2D, patients with interferon-γ response pathway gene mutations presented significantly increased interferon-γ receptor 2 expression, relative to those without mutation (p = 0.018). TP53 is critically involved in tumour progression, including DLBCL. Decitabine shows promising efficacy in treating patients with acute myeloid leukemia or myelodysplastic syndromes through targeting TP53 mutations.10 It is worth notifying that all five DLBCL patients with TP53 mutation achieved complete response and remained progression-free till last follow-up (Figure 3A). The possible structure-function relationship of TP53 was addressed using the crystal structure of the protein. TP53 K132R, F134C, R175H, G187fs, F212fs, R282W and E285K could disrupt DNA binding domain (Figure 3B). Moreover, significant elevation of peripheral CD3+T, CD3+CD4+T, CD3+CD8+T cells and serum interferon-γ were observed in mutant (MUT)-TP53 patients, as other DR-CHOP-responding patients (Figure 3C). To further determine the microenvironment influence of decitabine on MUT-TP53 DLBCL, SU-DHL-4TP53-R248Q, SU-DHL-4TP53-R273C, SU-DHL-4TP53-R175H and wild-type (WT)-TP53 SU-DHL-4TP53-WT cells were established. Upon treatment with decitabine (330 nM) for 5 days and doxorubicin (key cytotoxic agent of R-CHOP, 200 nM) for 2 days at clinically achievable concentrations,11 T-helper 1 cells were significantly increased in the co-culture system of MUT-TP53 cells (SU-DHL-4TP53-R248Q, SU-DHL-4TP53-R273C and SU-DHL-4TP53-R175H) with peripheral blood mononuclear cells (p < 0.001, Figure 3D), which was not observed in SU-DHL-4TP53-WT cells (p = 0.057, Figure 3E). As mechanism of action, T-helper 1 cells secrete interferon-γ and exhibit anti-tumour activities during cell-mediated adaptive immune response.9 Accordingly, significantly increased interferon-γ production was observed in all MUT-TP53 cells (p < 0.001), but not in WT-TP53 cells (p = 0.105) upon treatment with decitabine and doxorubicin (Figure 3D,E). Therefore, decitabine could modulate the tumour microenvironment of TP53-mutated DLBCL through enhancing T-helper 1-mediated anti-tumour response. In conclusion, DR-CHOP was effective and safe in newly diagnosed DLBCL patients. Benefit impact of DR-CHOP on the tumour microenvironment further provided clinical rationale of targeting DNA methylation as an important immunomodulatory strategy in treating DLBCL. We appreciate the effort of the physicians for enrolling patients and thank all the patients involved for allowing us to analyze their clinical data. This study was supported, in part, by research funding from the National Natural Science Foundation of China (grant numbers: 82130004, 81830007, 82170178, 81670176 and 82070204), Chang Jiang Scholars Program, Shanghai Municipal Education Commission Gaofeng Clinical Medicine Grant Support (grant numbers: 20152206 and 20152208), Clinical Research Plan of Shanghai Hospital Development Center (grant number: SHDC2020CR1032B), Multicenter Clinical Research Project by Shanghai Jiao Tong University School of Medicine (grant number: DLY201601), Collaborative Innovation Center of Systems Biomedicine, and the Samuel Waxman Cancer Research Foundation. The authors declare that they have no competing interests. National Natural Science Foundation of China, Grant Numbers: 82130004, 81830007, 82170178, 81670176 and 82070204; Chang Jiang Scholars Program; Shanghai Municipal Education Commission Gaofeng Clinical Medicine Grant Support, Grant Numbers: 20152206 and 20152208; Clinical Research Plan of Shanghai Hospital Development Center, Grant Number: SHDC2020CR1032B; Multicenter Clinical Research Project by Shanghai Jiao Tong University School of Medicine, Grant Number: DLY201601; Collaborative Innovation Center of Systems Biomedicine; Samuel Waxman Cancer Research Foundation Please note: The publisher is not responsible for the content or functionality of any supporting information supplied by the authors. Any queries (other than missing content) should be directed to the corresponding author for the article.