Addition of BTK inhibitor orelabrutinib to rituximab improved anti-tumor effects in B cell lymphoma

Hui Yu, Xing Wang, Jiao Li, Yingying Ye, Dedao Wang, Wei Fang, Lan Mi, Ning Ding, Xiaogan Wang, Yuqin Song, Jun Zhu, Hui Yu, Xing Wang, Jiao Li, Yingying Ye, Dedao Wang, Wei Fang, Lan Mi, Ning Ding, Xiaogan Wang, Yuqin Song, Jun Zhu

Abstract

Bruton tyrosine kinase (BTK) inhibitor ibrutinib has been validated as an effective drug to treat B cell malignancies. Combined therapies comprising ibrutinib and anti-CD20 antibodies like rituximab were designed as a backbone in many clinical trials. However, the off-target inhibition of ibrutinib on interleukin-2 (IL-2)-inducible T cell kinase (ITK) may reduce rituximab's antibody-dependent cellular cytotoxicity (ADCC) efficacy. Orelabrutinib (Orel), a novel BTK inhibitor, was designed with high selectivity to BTK. In our study, we demonstrated in preclinical models that orelabrutinib in combination with rituximab could preserve NK-cell-mediated ADCC induced by rituximab and enhanced the apoptosis of tumor cells in vitro. The addition of orelabrutinib to rituximab had produced promising combined anti-tumor effects in B cell lymphomas in vivo. Collectively, combination therapy of orelabrutinib with rituximab would benefit patients with B cell lymphoma, especially those with relapsed or refractory disease.

Keywords: B cell lymphoma; BTK inhibitor; anti-CD20 antibody; combined effects.

Conflict of interest statement

The authors declare no competing interests.

© 2021 The Authors.

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
Orelabrutinib inhibited B cell lymphoma cell proliferation in vitro (A) B cell lymphoma cell lines (TMD8, HBL-1, SU-DHL-6, WSU-NHL, Mino, Z138, and Raji) were treated with indicated concentrations of orelabrutinib (Orel) or ibrutinib (IBR) for 72 h. Cell viability was measured by Cell Titer-Glo luminescent cell viability assay. Data are shown as mean ± SD of three biological replicates. (B) Western blot analysis for phosphorylation levels of Bruton’s tyrosine kinase (BTK) and its related signaling pathway proteins in Z138 and HBL-1 cells after IBR or Orel treatment for 2 h. (C) The relative phosphorylation levels of signaling proteins from three biologically repeated experiments were quantified by measuring the relative intensity of phosphorylated bands to the β-actin bands. ∗p 

Figure 2

Orelabrutinib lacked off-target inhibition effect…

Figure 2

Orelabrutinib lacked off-target inhibition effect on cellular IL-2-inducible T cell kinase (ITK) compared…

Figure 2
Orelabrutinib lacked off-target inhibition effect on cellular IL-2-inducible T cell kinase (ITK) compared with ibrutinib (A and B) p-ITK and p-IκBα were assessed of proteins obtained from YT and NK-92 cells (two kinds of NK/T lymphoma cell lines expressing ITK) treated with ibrutinib or orelabrutinib for 2 h. PBMCs obtained from healthy adults were also used to verify the different effects of orelabrutinib and ibrutinib on ITK inhibition. After being pretreated with or without CD3/CD28-activating agent, PBMCs were treated with different concentrations of BTK inhibitors, orelabrutinib or ibrutinib. Proteins were extracted from pre-treated cells, and proteins of p-ITK and its associated downstream pathway were analyzed by western blot. These western blots were quantified from three biologically repeated tests. ∗p 

Figure 3

Orelabrutinib preserved rituximab-mediated cytotoxicity (A)…

Figure 3

Orelabrutinib preserved rituximab-mediated cytotoxicity (A) NK cells pretreated with 1 μmol/L BTK inhibitor,…

Figure 3
Orelabrutinib preserved rituximab-mediated cytotoxicity (A) NK cells pretreated with 1 μmol/L BTK inhibitor, ibrutinib, or Orel were co-cultured with B lymphoma cells at different E:T ratio in the presence of rituximab (RTX) at a concentration of 10 μg/mL or not. (B) Pretreated NK cells with 1 μmol/L ibrutinib or Orel were co-cultured with B lymphoma cells at 5:1 in the presence of 10 μg/mL rituximab or PBS. (C) Rituximab (10 μg/mL) or PBS was combined with the increasing concentration of Orel to evaluate tumor cell lysis induced by NK-cell-mediated antibody-dependent cell-mediated cytotoxicity (ADCC). Cell supernatant was collected after 4-h co-culture and then was measured for lactate dehydrogenase (LDH) with Non-Radioactive Cytotoxicity Assay. Each cell line was biologically tested for at least 3 times. ∗p 

Figure 4

Orelabrutinib enhanced apoptosis of tumor…

Figure 4

Orelabrutinib enhanced apoptosis of tumor cells induced by rituximab (A and B) B…

Figure 4
Orelabrutinib enhanced apoptosis of tumor cells induced by rituximab (A and B) B cell lymphoma cells (TMD8 and Z138) were treated with increasing concentration of ibrutinib or Orel for 48 h; after then, cells were stained with PI and Annexin V-FITC. Statistical analysis was performed using one-way ANOVA. Values from three independent experiments are presented as percentages of vehicle in mean ± SD. ∗p + cells were gated to analyze tumor apoptosis by flow cytometer. The column graph represents the percent of cell apoptosis for each group. Statistical analysis was performed using one-way ANOVA. Values of three biologically independent tests are presented as percentages of vehicle in mean ± SD. ∗p < 0.05 compared with vehicle group; ∗∗p < 0.01 compared with control group. #p < 0.05 compared with rituximab group; ##p < 0.01 compared with rituximab group.

Figure 5

Orelabrutinib combined with rituximab effectively…

Figure 5

Orelabrutinib combined with rituximab effectively inhibited tumor growth in animal models (A) CB.17/SCID…

Figure 5
Orelabrutinib combined with rituximab effectively inhibited tumor growth in animal models (A) CB.17/SCID mice were inoculated subcutaneously with 1 × 107 TMD8 cells in 0.1 mL PBS with Matrigel (1:1 ratio); n = 5 per group. (B) 1 mm3 patient tumor tissue was inoculated subcutaneously into the right flank of per mouse. n = 6 per group. Orelabrutinib (10 mg/kg, bid) and rituximab (200 μg/dose, weekly) were administered to the tumor-bearing mice when the tumor volume achieved 150 mm3. Body weight and tumor volume of mice were measured every other day during treatment. (C) Splenocytes of mice from different group were co-cultured with MCL cell line Z138 for 4 h. NK cell cytotoxic activity was then measured using the Nonradioactive Cytotoxicity Assay Kit. Each column represents the mean value of the triplicate experiments. (D) Hematoxylin and eosin (H&E) staining of tumor tissues from TMD8 cell-line-derived xenograft (CDX) tumor model and patient-derived xenograft (PDX) model. Scale bar: 70 μm. ∗p < 0.05 compared with control group, ∗∗p < 0.01 compared with control group, and ∗∗∗p < 0.001 compared with control group; #p < 0.05 compared with rituximab group and ###p < 0.001 compared with rituximab group.

Figure 6

Combined effects of orelabrutinib and…

Figure 6

Combined effects of orelabrutinib and rituximab in vivo (A and B) Apoptosis of…

Figure 6
Combined effects of orelabrutinib and rituximab in vivo (A and B) Apoptosis of tumor tissues from TMD8 tumor model was assessed by the terminal deoxynucleotidyl transferase nick-end-labeling (TUNEL) assay, and the nuclei were counterstained with DAPI. Representative images show apoptotic (fragmented) DNA (green staining) and the corresponding cell nuclei (blue) staining. Scale bar: 50 μm. Results are expressed as mean ± SD (n = 3 biologically independent samples). (C and D) Ki67, granzyme B, and NKp44 were assessed by immunohistochemistry of TMD8 tumor tissue. Scale bar: 60 μm. The data show the density of positivity cells for each section. Student’s t test was performed for statistical analysis. Values present as percentages of control group in mean ± SD (n = 3 biologically independent samples), ∗∗p 
All figures (7)
Similar articles
Cited by
References
    1. Herrera A.F. Noncellular immune therapies for non-Hodgkin lymphoma. Hematol. Oncol. Clin. North Am. 2019;33:707–725. - PubMed
    1. Sehn L.H., Donaldson J., Chhanabhai M., Fitzgerald C., Gill K., Klasa R., MacPherson N., O’Reilly S., Spinelli J.J., Sutherland J. Introduction of combined CHOP plus rituximab therapy dramatically improved outcome of diffuse large B-cell lymphoma in British Columbia. J. Clin. Oncol. 2005;23:5027–5033. - PubMed
    1. Hussain A., Yu L., Faryal R., Mohammad D.K., Mohamed A.J., Smith C.I. TEC family kinases in health and disease--loss-of-function of BTK and ITK and the gain-of-function fusions ITK-SYK and BTK-SYK. FEBS J. 2011;278:2001–2010. - PubMed
    1. Tomlinson M.G., Kane L.P., Su J., Kadlecek T.A., Mollenauer M.N., Weiss A. Expression and function of Tec, Itk, and Btk in lymphocytes: evidence for a unique role for Tec. Mol. Cell. Biol. 2004;24:2455–2466. - PMC - PubMed
    1. Davis R.E., Ngo V.N., Lenz G., Tolar P., Young R.M., Romesser P.B., Kohlhammer H., Lamy L., Zhao H., Yang Y. Chronic active B-cell-receptor signalling in diffuse large B-cell lymphoma. Nature. 2010;463:88–92. - PMC - PubMed
Show all 41 references
Related information
LinkOut - more resources
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 2
Figure 2
Orelabrutinib lacked off-target inhibition effect on cellular IL-2-inducible T cell kinase (ITK) compared with ibrutinib (A and B) p-ITK and p-IκBα were assessed of proteins obtained from YT and NK-92 cells (two kinds of NK/T lymphoma cell lines expressing ITK) treated with ibrutinib or orelabrutinib for 2 h. PBMCs obtained from healthy adults were also used to verify the different effects of orelabrutinib and ibrutinib on ITK inhibition. After being pretreated with or without CD3/CD28-activating agent, PBMCs were treated with different concentrations of BTK inhibitors, orelabrutinib or ibrutinib. Proteins were extracted from pre-treated cells, and proteins of p-ITK and its associated downstream pathway were analyzed by western blot. These western blots were quantified from three biologically repeated tests. ∗p 

Figure 3

Orelabrutinib preserved rituximab-mediated cytotoxicity (A)…

Figure 3

Orelabrutinib preserved rituximab-mediated cytotoxicity (A) NK cells pretreated with 1 μmol/L BTK inhibitor,…

Figure 3
Orelabrutinib preserved rituximab-mediated cytotoxicity (A) NK cells pretreated with 1 μmol/L BTK inhibitor, ibrutinib, or Orel were co-cultured with B lymphoma cells at different E:T ratio in the presence of rituximab (RTX) at a concentration of 10 μg/mL or not. (B) Pretreated NK cells with 1 μmol/L ibrutinib or Orel were co-cultured with B lymphoma cells at 5:1 in the presence of 10 μg/mL rituximab or PBS. (C) Rituximab (10 μg/mL) or PBS was combined with the increasing concentration of Orel to evaluate tumor cell lysis induced by NK-cell-mediated antibody-dependent cell-mediated cytotoxicity (ADCC). Cell supernatant was collected after 4-h co-culture and then was measured for lactate dehydrogenase (LDH) with Non-Radioactive Cytotoxicity Assay. Each cell line was biologically tested for at least 3 times. ∗p 

Figure 4

Orelabrutinib enhanced apoptosis of tumor…

Figure 4

Orelabrutinib enhanced apoptosis of tumor cells induced by rituximab (A and B) B…

Figure 4
Orelabrutinib enhanced apoptosis of tumor cells induced by rituximab (A and B) B cell lymphoma cells (TMD8 and Z138) were treated with increasing concentration of ibrutinib or Orel for 48 h; after then, cells were stained with PI and Annexin V-FITC. Statistical analysis was performed using one-way ANOVA. Values from three independent experiments are presented as percentages of vehicle in mean ± SD. ∗p + cells were gated to analyze tumor apoptosis by flow cytometer. The column graph represents the percent of cell apoptosis for each group. Statistical analysis was performed using one-way ANOVA. Values of three biologically independent tests are presented as percentages of vehicle in mean ± SD. ∗p < 0.05 compared with vehicle group; ∗∗p < 0.01 compared with control group. #p < 0.05 compared with rituximab group; ##p < 0.01 compared with rituximab group.

Figure 5

Orelabrutinib combined with rituximab effectively…

Figure 5

Orelabrutinib combined with rituximab effectively inhibited tumor growth in animal models (A) CB.17/SCID…

Figure 5
Orelabrutinib combined with rituximab effectively inhibited tumor growth in animal models (A) CB.17/SCID mice were inoculated subcutaneously with 1 × 107 TMD8 cells in 0.1 mL PBS with Matrigel (1:1 ratio); n = 5 per group. (B) 1 mm3 patient tumor tissue was inoculated subcutaneously into the right flank of per mouse. n = 6 per group. Orelabrutinib (10 mg/kg, bid) and rituximab (200 μg/dose, weekly) were administered to the tumor-bearing mice when the tumor volume achieved 150 mm3. Body weight and tumor volume of mice were measured every other day during treatment. (C) Splenocytes of mice from different group were co-cultured with MCL cell line Z138 for 4 h. NK cell cytotoxic activity was then measured using the Nonradioactive Cytotoxicity Assay Kit. Each column represents the mean value of the triplicate experiments. (D) Hematoxylin and eosin (H&E) staining of tumor tissues from TMD8 cell-line-derived xenograft (CDX) tumor model and patient-derived xenograft (PDX) model. Scale bar: 70 μm. ∗p < 0.05 compared with control group, ∗∗p < 0.01 compared with control group, and ∗∗∗p < 0.001 compared with control group; #p < 0.05 compared with rituximab group and ###p < 0.001 compared with rituximab group.

Figure 6

Combined effects of orelabrutinib and…

Figure 6

Combined effects of orelabrutinib and rituximab in vivo (A and B) Apoptosis of…

Figure 6
Combined effects of orelabrutinib and rituximab in vivo (A and B) Apoptosis of tumor tissues from TMD8 tumor model was assessed by the terminal deoxynucleotidyl transferase nick-end-labeling (TUNEL) assay, and the nuclei were counterstained with DAPI. Representative images show apoptotic (fragmented) DNA (green staining) and the corresponding cell nuclei (blue) staining. Scale bar: 50 μm. Results are expressed as mean ± SD (n = 3 biologically independent samples). (C and D) Ki67, granzyme B, and NKp44 were assessed by immunohistochemistry of TMD8 tumor tissue. Scale bar: 60 μm. The data show the density of positivity cells for each section. Student’s t test was performed for statistical analysis. Values present as percentages of control group in mean ± SD (n = 3 biologically independent samples), ∗∗p 
All figures (7)
Similar articles
Cited by
References
    1. Herrera A.F. Noncellular immune therapies for non-Hodgkin lymphoma. Hematol. Oncol. Clin. North Am. 2019;33:707–725. - PubMed
    1. Sehn L.H., Donaldson J., Chhanabhai M., Fitzgerald C., Gill K., Klasa R., MacPherson N., O’Reilly S., Spinelli J.J., Sutherland J. Introduction of combined CHOP plus rituximab therapy dramatically improved outcome of diffuse large B-cell lymphoma in British Columbia. J. Clin. Oncol. 2005;23:5027–5033. - PubMed
    1. Hussain A., Yu L., Faryal R., Mohammad D.K., Mohamed A.J., Smith C.I. TEC family kinases in health and disease--loss-of-function of BTK and ITK and the gain-of-function fusions ITK-SYK and BTK-SYK. FEBS J. 2011;278:2001–2010. - PubMed
    1. Tomlinson M.G., Kane L.P., Su J., Kadlecek T.A., Mollenauer M.N., Weiss A. Expression and function of Tec, Itk, and Btk in lymphocytes: evidence for a unique role for Tec. Mol. Cell. Biol. 2004;24:2455–2466. - PMC - PubMed
    1. Davis R.E., Ngo V.N., Lenz G., Tolar P., Young R.M., Romesser P.B., Kohlhammer H., Lamy L., Zhao H., Yang Y. Chronic active B-cell-receptor signalling in diffuse large B-cell lymphoma. Nature. 2010;463:88–92. - PMC - PubMed
Show all 41 references
Related information
LinkOut - more resources
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 3
Figure 3
Orelabrutinib preserved rituximab-mediated cytotoxicity (A) NK cells pretreated with 1 μmol/L BTK inhibitor, ibrutinib, or Orel were co-cultured with B lymphoma cells at different E:T ratio in the presence of rituximab (RTX) at a concentration of 10 μg/mL or not. (B) Pretreated NK cells with 1 μmol/L ibrutinib or Orel were co-cultured with B lymphoma cells at 5:1 in the presence of 10 μg/mL rituximab or PBS. (C) Rituximab (10 μg/mL) or PBS was combined with the increasing concentration of Orel to evaluate tumor cell lysis induced by NK-cell-mediated antibody-dependent cell-mediated cytotoxicity (ADCC). Cell supernatant was collected after 4-h co-culture and then was measured for lactate dehydrogenase (LDH) with Non-Radioactive Cytotoxicity Assay. Each cell line was biologically tested for at least 3 times. ∗p 

Figure 4

Orelabrutinib enhanced apoptosis of tumor…

Figure 4

Orelabrutinib enhanced apoptosis of tumor cells induced by rituximab (A and B) B…

Figure 4
Orelabrutinib enhanced apoptosis of tumor cells induced by rituximab (A and B) B cell lymphoma cells (TMD8 and Z138) were treated with increasing concentration of ibrutinib or Orel for 48 h; after then, cells were stained with PI and Annexin V-FITC. Statistical analysis was performed using one-way ANOVA. Values from three independent experiments are presented as percentages of vehicle in mean ± SD. ∗p + cells were gated to analyze tumor apoptosis by flow cytometer. The column graph represents the percent of cell apoptosis for each group. Statistical analysis was performed using one-way ANOVA. Values of three biologically independent tests are presented as percentages of vehicle in mean ± SD. ∗p < 0.05 compared with vehicle group; ∗∗p < 0.01 compared with control group. #p < 0.05 compared with rituximab group; ##p < 0.01 compared with rituximab group.

Figure 5

Orelabrutinib combined with rituximab effectively…

Figure 5

Orelabrutinib combined with rituximab effectively inhibited tumor growth in animal models (A) CB.17/SCID…

Figure 5
Orelabrutinib combined with rituximab effectively inhibited tumor growth in animal models (A) CB.17/SCID mice were inoculated subcutaneously with 1 × 107 TMD8 cells in 0.1 mL PBS with Matrigel (1:1 ratio); n = 5 per group. (B) 1 mm3 patient tumor tissue was inoculated subcutaneously into the right flank of per mouse. n = 6 per group. Orelabrutinib (10 mg/kg, bid) and rituximab (200 μg/dose, weekly) were administered to the tumor-bearing mice when the tumor volume achieved 150 mm3. Body weight and tumor volume of mice were measured every other day during treatment. (C) Splenocytes of mice from different group were co-cultured with MCL cell line Z138 for 4 h. NK cell cytotoxic activity was then measured using the Nonradioactive Cytotoxicity Assay Kit. Each column represents the mean value of the triplicate experiments. (D) Hematoxylin and eosin (H&E) staining of tumor tissues from TMD8 cell-line-derived xenograft (CDX) tumor model and patient-derived xenograft (PDX) model. Scale bar: 70 μm. ∗p < 0.05 compared with control group, ∗∗p < 0.01 compared with control group, and ∗∗∗p < 0.001 compared with control group; #p < 0.05 compared with rituximab group and ###p < 0.001 compared with rituximab group.

Figure 6

Combined effects of orelabrutinib and…

Figure 6

Combined effects of orelabrutinib and rituximab in vivo (A and B) Apoptosis of…

Figure 6
Combined effects of orelabrutinib and rituximab in vivo (A and B) Apoptosis of tumor tissues from TMD8 tumor model was assessed by the terminal deoxynucleotidyl transferase nick-end-labeling (TUNEL) assay, and the nuclei were counterstained with DAPI. Representative images show apoptotic (fragmented) DNA (green staining) and the corresponding cell nuclei (blue) staining. Scale bar: 50 μm. Results are expressed as mean ± SD (n = 3 biologically independent samples). (C and D) Ki67, granzyme B, and NKp44 were assessed by immunohistochemistry of TMD8 tumor tissue. Scale bar: 60 μm. The data show the density of positivity cells for each section. Student’s t test was performed for statistical analysis. Values present as percentages of control group in mean ± SD (n = 3 biologically independent samples), ∗∗p 
All figures (7)
Similar articles
Cited by
References
    1. Herrera A.F. Noncellular immune therapies for non-Hodgkin lymphoma. Hematol. Oncol. Clin. North Am. 2019;33:707–725. - PubMed
    1. Sehn L.H., Donaldson J., Chhanabhai M., Fitzgerald C., Gill K., Klasa R., MacPherson N., O’Reilly S., Spinelli J.J., Sutherland J. Introduction of combined CHOP plus rituximab therapy dramatically improved outcome of diffuse large B-cell lymphoma in British Columbia. J. Clin. Oncol. 2005;23:5027–5033. - PubMed
    1. Hussain A., Yu L., Faryal R., Mohammad D.K., Mohamed A.J., Smith C.I. TEC family kinases in health and disease--loss-of-function of BTK and ITK and the gain-of-function fusions ITK-SYK and BTK-SYK. FEBS J. 2011;278:2001–2010. - PubMed
    1. Tomlinson M.G., Kane L.P., Su J., Kadlecek T.A., Mollenauer M.N., Weiss A. Expression and function of Tec, Itk, and Btk in lymphocytes: evidence for a unique role for Tec. Mol. Cell. Biol. 2004;24:2455–2466. - PMC - PubMed
    1. Davis R.E., Ngo V.N., Lenz G., Tolar P., Young R.M., Romesser P.B., Kohlhammer H., Lamy L., Zhao H., Yang Y. Chronic active B-cell-receptor signalling in diffuse large B-cell lymphoma. Nature. 2010;463:88–92. - PMC - PubMed
Show all 41 references
Related information
LinkOut - more resources
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 4
Figure 4
Orelabrutinib enhanced apoptosis of tumor cells induced by rituximab (A and B) B cell lymphoma cells (TMD8 and Z138) were treated with increasing concentration of ibrutinib or Orel for 48 h; after then, cells were stained with PI and Annexin V-FITC. Statistical analysis was performed using one-way ANOVA. Values from three independent experiments are presented as percentages of vehicle in mean ± SD. ∗p + cells were gated to analyze tumor apoptosis by flow cytometer. The column graph represents the percent of cell apoptosis for each group. Statistical analysis was performed using one-way ANOVA. Values of three biologically independent tests are presented as percentages of vehicle in mean ± SD. ∗p < 0.05 compared with vehicle group; ∗∗p < 0.01 compared with control group. #p < 0.05 compared with rituximab group; ##p < 0.01 compared with rituximab group.
Figure 5
Figure 5
Orelabrutinib combined with rituximab effectively inhibited tumor growth in animal models (A) CB.17/SCID mice were inoculated subcutaneously with 1 × 107 TMD8 cells in 0.1 mL PBS with Matrigel (1:1 ratio); n = 5 per group. (B) 1 mm3 patient tumor tissue was inoculated subcutaneously into the right flank of per mouse. n = 6 per group. Orelabrutinib (10 mg/kg, bid) and rituximab (200 μg/dose, weekly) were administered to the tumor-bearing mice when the tumor volume achieved 150 mm3. Body weight and tumor volume of mice were measured every other day during treatment. (C) Splenocytes of mice from different group were co-cultured with MCL cell line Z138 for 4 h. NK cell cytotoxic activity was then measured using the Nonradioactive Cytotoxicity Assay Kit. Each column represents the mean value of the triplicate experiments. (D) Hematoxylin and eosin (H&E) staining of tumor tissues from TMD8 cell-line-derived xenograft (CDX) tumor model and patient-derived xenograft (PDX) model. Scale bar: 70 μm. ∗p < 0.05 compared with control group, ∗∗p < 0.01 compared with control group, and ∗∗∗p < 0.001 compared with control group; #p < 0.05 compared with rituximab group and ###p < 0.001 compared with rituximab group.
Figure 6
Figure 6
Combined effects of orelabrutinib and rituximab in vivo (A and B) Apoptosis of tumor tissues from TMD8 tumor model was assessed by the terminal deoxynucleotidyl transferase nick-end-labeling (TUNEL) assay, and the nuclei were counterstained with DAPI. Representative images show apoptotic (fragmented) DNA (green staining) and the corresponding cell nuclei (blue) staining. Scale bar: 50 μm. Results are expressed as mean ± SD (n = 3 biologically independent samples). (C and D) Ki67, granzyme B, and NKp44 were assessed by immunohistochemistry of TMD8 tumor tissue. Scale bar: 60 μm. The data show the density of positivity cells for each section. Student’s t test was performed for statistical analysis. Values present as percentages of control group in mean ± SD (n = 3 biologically independent samples), ∗∗p 
All figures (7)

References

    1. Herrera A.F. Noncellular immune therapies for non-Hodgkin lymphoma. Hematol. Oncol. Clin. North Am. 2019;33:707–725.
    1. Sehn L.H., Donaldson J., Chhanabhai M., Fitzgerald C., Gill K., Klasa R., MacPherson N., O’Reilly S., Spinelli J.J., Sutherland J. Introduction of combined CHOP plus rituximab therapy dramatically improved outcome of diffuse large B-cell lymphoma in British Columbia. J. Clin. Oncol. 2005;23:5027–5033.
    1. Hussain A., Yu L., Faryal R., Mohammad D.K., Mohamed A.J., Smith C.I. TEC family kinases in health and disease--loss-of-function of BTK and ITK and the gain-of-function fusions ITK-SYK and BTK-SYK. FEBS J. 2011;278:2001–2010.
    1. Tomlinson M.G., Kane L.P., Su J., Kadlecek T.A., Mollenauer M.N., Weiss A. Expression and function of Tec, Itk, and Btk in lymphocytes: evidence for a unique role for Tec. Mol. Cell. Biol. 2004;24:2455–2466.
    1. Davis R.E., Ngo V.N., Lenz G., Tolar P., Young R.M., Romesser P.B., Kohlhammer H., Lamy L., Zhao H., Yang Y. Chronic active B-cell-receptor signalling in diffuse large B-cell lymphoma. Nature. 2010;463:88–92.
    1. Advani R.H., Buggy J.J., Sharman J.P., Smith S.M., Boyd T.E., Grant B., Kolibaba K.S., Furman R.R., Rodriguez S., Chang B.Y. Bruton tyrosine kinase inhibitor ibrutinib (PCI-32765) has significant activity in patients with relapsed/refractory B-cell malignancies. J. Clin. Oncol. 2013;31:88–94.
    1. Burger J.A., Buggy J.J. Bruton tyrosine kinase inhibitor ibrutinib (PCI-32765) Leuk. Lymphoma. 2013;54:2385–2391.
    1. Byrd J.C., Furman R.R., Coutre S.E., Flinn I.W., Burger J.A., Blum K.A., Grant B., Sharman J.P., Coleman M., Wierda W.G. Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia. N. Engl. J. Med. 2013;369:32–42.
    1. Wang M.L., Rule S., Martin P., Goy A., Auer R., Kahl B.S., Jurczak W., Advani R.H., Romaguera J.E., Williams M.E. Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma. N. Engl. J. Med. 2013;369:507–516.
    1. Brower V. Ibrutinib promising in subtype of DLBCL. Lancet Oncol. 2015;16:e428.
    1. Burger J.A., Tedeschi A., Barr P.M., Robak T., Owen C., Ghia P., Bairey O., Hillmen P., Bartlett N.L., Li J., RESONATE-2 Investigators Ibrutinib as initial therapy for patients with chronic lymphocytic leukemia. N. Engl. J. Med. 2015;373:2425–2437.
    1. Treon S.P., Tripsas C.K., Meid K., Warren D., Varma G., Green R., Argyropoulos K.V., Yang G., Cao Y., Xu L. Ibrutinib in previously treated Waldenström’s macroglobulinemia. N. Engl. J. Med. 2015;372:1430–1440.
    1. Dubovsky J.A., Beckwith K.A., Natarajan G., Woyach J.A., Jaglowski S., Zhong Y., Hessler J.D., Liu T.M., Chang B.Y., Larkin K.M. Ibrutinib is an irreversible molecular inhibitor of ITK driving a Th1-selective pressure in T lymphocytes. Blood. 2013;122:2539–2549.
    1. Younes A., Thieblemont C., Morschhauser F., Flinn I., Friedberg J.W., Amorim S., Hivert B., Westin J., Vermeulen J., Bandyopadhyay N. Combination of ibrutinib with rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) for treatment-naive patients with CD20-positive B-cell non-Hodgkin lymphoma: a non-randomised, phase 1b study. Lancet Oncol. 2014;15:1019–1026.
    1. Kohrt H.E., Sagiv-Barfi I., Rafiq S., Herman S.E., Butchar J.P., Cheney C., Zhang X., Buggy J.J., Muthusamy N., Levy R. Ibrutinib antagonizes rituximab-dependent NK cell-mediated cytotoxicity. Blood. 2014;123:1957–1960.
    1. Burger J.A., Keating M.J., Wierda W.G., Hartmann E., Hoellenriegel J., Rosin N.Y., de Weerdt I., Jeyakumar G., Ferrajoli A., Cardenas-Turanzas M. Safety and activity of ibrutinib plus rituximab for patients with high-risk chronic lymphocytic leukaemia: a single-arm, phase 2 study. Lancet Oncol. 2014;15:1090–1099.
    1. Dhillon S. Orelabrutinib: first approval. Drugs. 2021;81:503–507.
    1. Song Y., Song Y., Liu L., Zhang M., Li Z., Ji C., Xu W., Liu T., Xu B., Wang X. Safety and efficacy of orelabrutinib monotherapy in Chinese patients with relapsed or refractory mantle cell lymphoma: a multicenter, open-label, phase II study. Blood. 2019;134:755.
    1. Clynes R.A., Towers T.L., Presta L.G., Ravetch J.V. Inhibitory Fc receptors modulate in vivo cytotoxicity against tumor targets. Nat. Med. 2000;6:443–446.
    1. Bowles J.A., Weiner G.J. CD16 polymorphisms and NK activation induced by monoclonal antibody-coated target cells. J. Immunol. Methods. 2005;304:88–99.
    1. Herman S.E., Mustafa R.Z., Gyamfi J.A., Pittaluga S., Chang S., Chang B., Farooqui M., Wiestner A. Ibrutinib inhibits BCR and NF-κB signaling and reduces tumor proliferation in tissue-resident cells of patients with CLL. Blood. 2014;123:3286–3295.
    1. Zhao X., Bodo J., Sun D., Durkin L., Lin J., Smith M.R., Hsi E.D. Combination of ibrutinib with ABT-199: synergistic effects on proliferation inhibition and apoptosis in mantle cell lymphoma cells through perturbation of BTK, AKT and BCL2 pathways. Br. J. Haematol. 2015;168:765–768.
    1. Yasuhiro T., Sawada W., Klein C., Kozaki R., Hotta S., Yoshizawa T. Anti-tumor efficacy study of the Bruton’s tyrosine kinase (BTK) inhibitor, ONO/GS-4059, in combination with the glycoengineered type II anti-CD20 monoclonal antibody obinutuzumab (GA101) demonstrates superior in vivo efficacy compared to ONO/GS-4059 in combination with rituximab. Leuk. Lymphoma. 2017;58:699–707.
    1. Reiff S.D., Muhowski E.M., Guinn D., Lehman A., Fabian C.A., Cheney C., Mantel R., Smith L., Johnson A.J., Young W.B. Noncovalent inhibition of C481S Bruton tyrosine kinase by GDC-0853: a new treatment strategy for ibrutinib-resistant CLL. Blood. 2018;132:1039–1049.
    1. Zhou X., Hu W., Qin X. The role of complement in the mechanism of action of rituximab for B-cell lymphoma: implications for therapy. Oncologist. 2008;13:954–966.
    1. Bondza S., Marosan A., Kara S., Lösing J., Peipp M., Nimmerjahn F., Buijs J., Lux A. Complement-dependent activity of CD20-specific IgG correlates with bivalent antigen binding and C1q binding strength. Front. Immunol. 2021;11:609941.
    1. Chang B.Y., Francesco M., De Rooij M.F., Magadala P., Steggerda S.M., Huang M.M., Kuil A., Herman S.E., Chang S., Pals S.T. Egress of CD19(+)CD5(+) cells into peripheral blood following treatment with the Bruton tyrosine kinase inhibitor ibrutinib in mantle cell lymphoma patients. Blood. 2013;122:2412–2424.
    1. Bojarczuk K., Siernicka M., Dwojak M., Bobrowicz M., Pyrzynska B., Gaj P., Karp M., Giannopoulos K., Efremov D.G., Fauriat C. B-cell receptor pathway inhibitors affect CD20 levels and impair antitumor activity of anti-CD20 monoclonal antibodies. Leukemia. 2014;28:1163–1167.
    1. Bobrowicz M., Dwojak M., Pyrzynska B., Stachura J., Muchowicz A., Berthel E., Dalla-Venezia N., Kozikowski M., Siernicka M., Miazek N. HDAC6 inhibition upregulates CD20 levels and increases the efficacy of anti-CD20 monoclonal antibodies. Blood. 2017;130:1628–1638.
    1. Rougé L., Chiang N., Steffek M., Kugel C., Croll T.I., Tam C., Estevez A., Arthur C.P., Koth C.M., Ciferri C. Structure of CD20 in complex with the therapeutic monoclonal antibody rituximab. Science. 2020;367:1224–1230.
    1. Marshall M.J.E., Stopforth R.J., Cragg M.S. Therapeutic antibodies: what have we learnt from targeting CD20 and where are we going? Front. Immunol. 2017;8:1245.
    1. Tarazona R., Campos C., Pera A., Sanchez-Correa B., Solana R. Flow cytometry analysis of NK cell phenotype and function in aging. Methods Mol. Biol. 2015;1343:9–18.
    1. Böttcher J.P., Bonavita E., Chakravarty P., Blees H., Cabeza-Cabrerizo M., Sammicheli S., Rogers N.C., Sahai E., Zelenay S., Reis E Sousa C. NK cells stimulate recruitment of cDC1 into the tumor microenvironment promoting cancer immune control. Cell. 2018;172:1022–1037.e14.
    1. Sagiv-Barfi I., Kohrt H.E., Czerwinski D.K., Ng P.P., Chang B.Y., Levy R. Therapeutic antitumor immunity by checkpoint blockade is enhanced by ibrutinib, an inhibitor of both BTK and ITK. Proc. Natl. Acad. Sci. USA. 2015;112:E966–E972.
    1. Vincent-Fabert C., Roland L., Zimber-Strobl U., Feuillard J., Faumont N. Pre-clinical blocking of PD-L1 molecule, which expression is down regulated by NF-κB, JAK1/JAK2 and BTK inhibitors, induces regression of activated B-cell lymphoma. Cell Commun. Signal. 2019;17:89.
    1. Byrd J.C., Harrington B., O’Brien S., Jones J.A., Schuh A., Devereux S., Chaves J., Wierda W.G., Awan F.T., Brown J.R. Acalabrutinib (ACP-196) in relapsed chronic lymphocytic leukemia. N. Engl. J. Med. 2016;374:323–332.
    1. Ping L., Ding N., Shi Y., Feng L., Li J., Liu Y., Lin Y., Shi C., Wang X., Pan Z. The Bruton’s tyrosine kinase inhibitor ibrutinib exerts immunomodulatory effects through regulation of tumor-infiltrating macrophages. Oncotarget. 2017;8:39218–39229.
    1. Long M., Beckwith K., Do P., Mundy B.L., Gordon A., Lehman A.M., Maddocks K.J., Cheney C., Jones J.A., Flynn J.M. Ibrutinib treatment improves T cell number and function in CLL patients. J. Clin. Invest. 2017;127:3052–3064.
    1. Kondo K., Shaim H., Thompson P.A., Burger J.A., Keating M., Estrov Z., Harris D., Kim E., Ferrajoli A., Daher M. Ibrutinib modulates the immunosuppressive CLL microenvironment through STAT3-mediated suppression of regulatory B-cell function and inhibition of the PD-1/PD-L1 pathway. Leukemia. 2018;32:960–970.
    1. Li J., Wang X., Xie Y., Ying Z., Liu W., Ping L., Zhang C., Pan Z., Ding N., Song Y., Zhu J. The mTOR kinase inhibitor everolimus synergistically enhances the anti-tumor effect of the Bruton’s tyrosine kinase (BTK) inhibitor PLS-123 on Mantle cell lymphoma. Int. J. Cancer. 2018;142:202–213.
    1. Ashizawa T., Iizuka A., Nonomura C., Kondou R., Maeda C., Miyata H., Sugino T., Mitsuya K., Hayashi N., Nakasu Y. Antitumor effect of programmed death-1 (PD-1) blockade in humanized the NOG-MHC double knockout mouse. Clin. Cancer Res. 2017;23:149–158.

Source: PubMed

3
Abonner