Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2024

Open Access 01-12-2024 | Melanoma | Research

Targeting HDAC6 improves anti-CD47 immunotherapy

Authors: Maria Gracia-Hernandez, Ashutosh S. Yende, Nithya Gajendran, Zubaydah Alahmadi, Xintang Li, Zuleima Munoz, Karen Tan, Satish Noonepalle, Maho Shibata, Alejandro Villagra

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2024

Login to get access

Abstract

Background

Cancer cells can overexpress CD47, an innate immune checkpoint that prevents phagocytosis upon interaction with signal regulatory protein alpha (SIRPα) expressed in macrophages and other myeloid cells. Several clinical trials have reported that CD47 blockade reduces tumor growth in hematological malignancies. However, CD47 blockade has shown modest results in solid tumors, including melanoma. Our group has demonstrated that histone deacetylase 6 inhibitors (HDAC6is) have immunomodulatory properties, such as controlling macrophage phenotype and inflammatory properties. However, the molecular and cellular mechanisms controlling these processes are not fully understood. In this study, we evaluated the role of HDAC6 in regulating the CD47/SIRPα axis and phagocytosis in macrophages.

Methods

We tested the role of HDAC6is, especially Nexturastat A, in regulating macrophage phenotype and phagocytic function using bone marrow-derived macrophages and macrophage cell lines. The modulation of the CD47/SIRPα axis and phagocytosis by HDAC6is was investigated using murine and human melanoma cell lines and macrophages. Phagocytosis was evaluated via coculture assays of macrophages and melanoma cells by flow cytometry and immunofluorescence. Lastly, to evaluate the antitumor activity of Nexturastat A in combination with anti-CD47 or anti-SIRPα antibodies, we performed in vivo studies using the SM1 and/or B16F10 melanoma mouse models.

Results

We observed that HDAC6is enhanced the phenotype of antitumoral M1 macrophages while decreasing the protumoral M2 phenotype. In addition, HDAC6 inhibition diminished the expression of SIRPα, increased the expression of other pro-phagocytic signals in macrophages, and downregulated CD47 expression in mouse and human melanoma cells. This regulatory role on the CD47/SIRPα axis translated into enhanced antitumoral phagocytic capacity of macrophages treated with Nexturastat A and anti-CD47. We also observed that the systemic administration of HDAC6i enhanced the in vivo antitumor activity of anti-CD47 blockade in melanoma by modulating macrophage and natural killer cells in the tumor microenvironment. However, Nexturastat A did not enhance the antitumor activity of anti-SIRPα despite its modulation of macrophage populations in the SM1 tumor microenvironment.

Conclusions

Our results demonstrate the critical regulatory role of HDAC6 in phagocytosis and innate immunity for the first time, further underscoring the use of these inhibitors to potentiate CD47 immune checkpoint blockade therapeutic strategies.
Appendix
Available only for authorised users
Literature
1.
go back to reference Strome SE, et al. B7-H1 blockade augments adoptive T-cell immunotherapy for squamous cell carcinoma. Cancer Res. 2003;63:6501–5.PubMed Strome SE, et al. B7-H1 blockade augments adoptive T-cell immunotherapy for squamous cell carcinoma. Cancer Res. 2003;63:6501–5.PubMed
2.
go back to reference Iwai Y, et al. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci. 2002;99:12293–7.ADSPubMedPubMedCentralCrossRef Iwai Y, et al. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci. 2002;99:12293–7.ADSPubMedPubMedCentralCrossRef
3.
go back to reference Dobry AS, et al. Management of metastatic melanoma: improved survival in a national cohort following the approvals of checkpoint blockade immunotherapies and targeted therapies. Cancer Immunol Immunother CII. 2018;67:1833–44.PubMedCrossRef Dobry AS, et al. Management of metastatic melanoma: improved survival in a national cohort following the approvals of checkpoint blockade immunotherapies and targeted therapies. Cancer Immunol Immunother CII. 2018;67:1833–44.PubMedCrossRef
4.
go back to reference Huang AC, Zappasodi R. A decade of checkpoint blockade immunotherapy in melanoma: understanding the molecular basis for immune sensitivity and resistance. Nat Immunol. 2022;23:660–70.PubMedPubMedCentralCrossRef Huang AC, Zappasodi R. A decade of checkpoint blockade immunotherapy in melanoma: understanding the molecular basis for immune sensitivity and resistance. Nat Immunol. 2022;23:660–70.PubMedPubMedCentralCrossRef
5.
go back to reference Wolchok JD, et al. Long-term outcomes with Nivolumab Plus Ipilimumab or Nivolumab alone Versus Ipilimumab in patients with Advanced Melanoma. J Clin Oncol off J Am Soc Clin Oncol. 2022;40:127–37.CrossRef Wolchok JD, et al. Long-term outcomes with Nivolumab Plus Ipilimumab or Nivolumab alone Versus Ipilimumab in patients with Advanced Melanoma. J Clin Oncol off J Am Soc Clin Oncol. 2022;40:127–37.CrossRef
7.
8.
go back to reference Matlung HL, Szilagyi K, Barclay NA, van den Berg TK. The CD47-SIRPα signaling axis as an innate immune checkpoint in cancer. Immunol Rev. 2017;276:145–64.PubMedCrossRef Matlung HL, Szilagyi K, Barclay NA, van den Berg TK. The CD47-SIRPα signaling axis as an innate immune checkpoint in cancer. Immunol Rev. 2017;276:145–64.PubMedCrossRef
9.
go back to reference Gardai SJ, et al. Cell-surface calreticulin initiates clearance of viable or Apoptotic Cells through trans-activation of LRP on the Phagocyte. Cell. 2005;123:321–34.PubMedCrossRef Gardai SJ, et al. Cell-surface calreticulin initiates clearance of viable or Apoptotic Cells through trans-activation of LRP on the Phagocyte. Cell. 2005;123:321–34.PubMedCrossRef
10.
go back to reference Oldenborg P-A, Gresham HD, Lindberg FP. Cd47-Signal Regulatory protein α (Sirpα) regulates Fcγ and complement receptor–mediated phagocytosis. J Exp Med. 2001;193:855–62.PubMedPubMedCentralCrossRef Oldenborg P-A, Gresham HD, Lindberg FP. Cd47-Signal Regulatory protein α (Sirpα) regulates Fcγ and complement receptor–mediated phagocytosis. J Exp Med. 2001;193:855–62.PubMedPubMedCentralCrossRef
11.
go back to reference Koga N, et al. Clinical significance of signal regulatory protein alpha (SIRPα) expression in esophageal squamous cell carcinoma. Cancer Sci. 2021;112:3018–28.PubMedPubMedCentralCrossRef Koga N, et al. Clinical significance of signal regulatory protein alpha (SIRPα) expression in esophageal squamous cell carcinoma. Cancer Sci. 2021;112:3018–28.PubMedPubMedCentralCrossRef
13.
go back to reference Kuo TC, et al. Targeting the myeloid checkpoint receptor SIRPα potentiates innate and adaptive immune responses to promote anti-tumor activity. J Hematol Oncol J Hematol Oncol. 2020;13:160.PubMedCrossRef Kuo TC, et al. Targeting the myeloid checkpoint receptor SIRPα potentiates innate and adaptive immune responses to promote anti-tumor activity. J Hematol Oncol J Hematol Oncol. 2020;13:160.PubMedCrossRef
15.
go back to reference Zhang W et al. Advances in Anti-tumor treatments targeting the CD47/SIRPα Axis. Front Immunol 11, (2020). Zhang W et al. Advances in Anti-tumor treatments targeting the CD47/SIRPα Axis. Front Immunol 11, (2020).
16.
go back to reference Jiang Z, Sun H, Yu J, Tian W, Song Y. Targeting CD47 for cancer immunotherapy. J Hematol Oncol J Hematol Oncol. 2021;14:180.PubMedCrossRef Jiang Z, Sun H, Yu J, Tian W, Song Y. Targeting CD47 for cancer immunotherapy. J Hematol Oncol J Hematol Oncol. 2021;14:180.PubMedCrossRef
17.
go back to reference Maute R, Xu J, Weissman IL. CD47–SIRPα-targeted therapeutics: status and prospects. Immuno-Oncol Technol. 2022;13:100070.CrossRef Maute R, Xu J, Weissman IL. CD47–SIRPα-targeted therapeutics: status and prospects. Immuno-Oncol Technol. 2022;13:100070.CrossRef
18.
go back to reference Sockolosky JT et al. Durable antitumor responses to CD47 blockade require adaptive immune stimulation. Proc. Natl. Acad. Sci. U. S. A 113, E2646–E2654 (2016). Sockolosky JT et al. Durable antitumor responses to CD47 blockade require adaptive immune stimulation. Proc. Natl. Acad. Sci. U. S. A 113, E2646–E2654 (2016).
20.
go back to reference Woan KV, et al. Targeting histone deacetylase 6 mediates a dual anti-melanoma effect: enhanced antitumor immunity and impaired cell proliferation. Mol Oncol. 2015;9:1447–57.PubMedPubMedCentralCrossRef Woan KV, et al. Targeting histone deacetylase 6 mediates a dual anti-melanoma effect: enhanced antitumor immunity and impaired cell proliferation. Mol Oncol. 2015;9:1447–57.PubMedPubMedCentralCrossRef
21.
go back to reference Knox T, et al. Selective HDAC6 inhibitors improve anti-PD-1 immune checkpoint blockade therapy by decreasing the anti-inflammatory phenotype of macrophages and down-regulation of immunosuppressive proteins in tumor cells. Sci Rep. 2019;9:6136.ADSPubMedPubMedCentralCrossRef Knox T, et al. Selective HDAC6 inhibitors improve anti-PD-1 immune checkpoint blockade therapy by decreasing the anti-inflammatory phenotype of macrophages and down-regulation of immunosuppressive proteins in tumor cells. Sci Rep. 2019;9:6136.ADSPubMedPubMedCentralCrossRef
22.
go back to reference Banik D, et al. HDAC6 plays a noncanonical role in the regulation of Antitumor Immune responses, dissemination, and invasiveness of breast Cancer. Cancer Res. 2020;80:3649–62.PubMedPubMedCentralCrossRef Banik D, et al. HDAC6 plays a noncanonical role in the regulation of Antitumor Immune responses, dissemination, and invasiveness of breast Cancer. Cancer Res. 2020;80:3649–62.PubMedPubMedCentralCrossRef
23.
go back to reference Laengle J, et al. Histone deacetylase inhibitors valproic acid and vorinostat enhance trastuzumab-mediated antibody-dependent cell-mediated phagocytosis. J Immunother Cancer. 2020;8:e000195.PubMedPubMedCentralCrossRef Laengle J, et al. Histone deacetylase inhibitors valproic acid and vorinostat enhance trastuzumab-mediated antibody-dependent cell-mediated phagocytosis. J Immunother Cancer. 2020;8:e000195.PubMedPubMedCentralCrossRef
24.
go back to reference Hirano M, et al. Histone deacetylase inhibitors with or without AKT Inhibition potentially increase the efficacy of Daratumumab in multiple Myeloma by enhancing the antibody-dependent cell-mediated and complement-dependent cytotoxicity as well as apoptosis. Blood. 2018;132:4435.CrossRef Hirano M, et al. Histone deacetylase inhibitors with or without AKT Inhibition potentially increase the efficacy of Daratumumab in multiple Myeloma by enhancing the antibody-dependent cell-mediated and complement-dependent cytotoxicity as well as apoptosis. Blood. 2018;132:4435.CrossRef
27.
go back to reference Cheng F, et al. A novel role for Histone Deacetylase 6 in the regulation of the Tolerogenic STAT3/IL-10 pathway in APCs. J Immunol. 2014;193:2850–62.PubMedCrossRef Cheng F, et al. A novel role for Histone Deacetylase 6 in the regulation of the Tolerogenic STAT3/IL-10 pathway in APCs. J Immunol. 2014;193:2850–62.PubMedCrossRef
28.
go back to reference Cheng F, et al. Epigenetic modulation of STAT3 by Histone Deacetylase 6 (HDAC6) regulates IL-10 gene expression and Immune Tolerance mediated by Antigen-presenting cells (APCs). Blood. 2011;118:519–9.CrossRef Cheng F, et al. Epigenetic modulation of STAT3 by Histone Deacetylase 6 (HDAC6) regulates IL-10 gene expression and Immune Tolerance mediated by Antigen-presenting cells (APCs). Blood. 2011;118:519–9.CrossRef
29.
go back to reference Tavares MT, et al. Synthesis and Pharmacological Evaluation of Selective Histone Deacetylase 6 inhibitors in Melanoma models. ACS Med Chem Lett. 2017;8:1031–6.PubMedPubMedCentralCrossRef Tavares MT, et al. Synthesis and Pharmacological Evaluation of Selective Histone Deacetylase 6 inhibitors in Melanoma models. ACS Med Chem Lett. 2017;8:1031–6.PubMedPubMedCentralCrossRef
30.
go back to reference Jayasingam SD et al. Evaluating the polarization of Tumor-Associated macrophages into M1 and M2 phenotypes in Human Cancer tissue: technicalities and challenges in Routine Clinical Practice. Front Oncol 9, (2020). Jayasingam SD et al. Evaluating the polarization of Tumor-Associated macrophages into M1 and M2 phenotypes in Human Cancer tissue: technicalities and challenges in Routine Clinical Practice. Front Oncol 9, (2020).
31.
go back to reference Kovacsovics-Bankowski M, Rock KL. Presentation of exogenous antigens by macrophages: analysis of major histocompatibility complex class I and II presentation and regulation by cytokines. Eur J Immunol. 1994;24:2421–8.PubMedCrossRef Kovacsovics-Bankowski M, Rock KL. Presentation of exogenous antigens by macrophages: analysis of major histocompatibility complex class I and II presentation and regulation by cytokines. Eur J Immunol. 1994;24:2421–8.PubMedCrossRef
32.
go back to reference Chen R-F, Wang L, Cheng J-T, Yang KD. Induction of IFNα or IL-12 depends on differentiation of THP-1 cells in dengue infections without and with antibody enhancement. BMC Infect Dis. 2012;12:340.PubMedPubMedCentralCrossRef Chen R-F, Wang L, Cheng J-T, Yang KD. Induction of IFNα or IL-12 depends on differentiation of THP-1 cells in dengue infections without and with antibody enhancement. BMC Infect Dis. 2012;12:340.PubMedPubMedCentralCrossRef
33.
go back to reference Venkatraman S, Meller J, Hongeng S, Tohtong R, Chutipongtanate S. Transcriptional regulation of Cancer Immune checkpoints: emerging strategies for Immunotherapy. Vaccines. 2020;8:735.PubMedPubMedCentralCrossRef Venkatraman S, Meller J, Hongeng S, Tohtong R, Chutipongtanate S. Transcriptional regulation of Cancer Immune checkpoints: emerging strategies for Immunotherapy. Vaccines. 2020;8:735.PubMedPubMedCentralCrossRef
34.
go back to reference Lin Y, et al. Notch Signaling modulates macrophage polarization and Phagocytosis through Direct Suppression of Signal Regulatory Protein α expression. Front Immunol. 2018;9:1744.PubMedPubMedCentralCrossRef Lin Y, et al. Notch Signaling modulates macrophage polarization and Phagocytosis through Direct Suppression of Signal Regulatory Protein α expression. Front Immunol. 2018;9:1744.PubMedPubMedCentralCrossRef
35.
37.
go back to reference Yanagita T et al. Anti-SIRPα antibodies as a potential new tool for cancer immunotherapy. JCI Insight 2, e89140. Yanagita T et al. Anti-SIRPα antibodies as a potential new tool for cancer immunotherapy. JCI Insight 2, e89140.
40.
go back to reference Gupta A, Taslim C, Tullius BP, Cripe TP. Therapeutic modulation of the CD47-SIRPα axis in the pediatric tumor microenvironment: working up an appetite. Cancer Drug Resist. 2020;3:550–62.PubMedPubMedCentral Gupta A, Taslim C, Tullius BP, Cripe TP. Therapeutic modulation of the CD47-SIRPα axis in the pediatric tumor microenvironment: working up an appetite. Cancer Drug Resist. 2020;3:550–62.PubMedPubMedCentral
41.
go back to reference Mosely SIS, et al. Rational selection of Syngeneic Preclinical Tumor models for Immunotherapeutic Drug Discovery. Cancer Immunol Res. 2017;5:29–41.PubMedCrossRef Mosely SIS, et al. Rational selection of Syngeneic Preclinical Tumor models for Immunotherapeutic Drug Discovery. Cancer Immunol Res. 2017;5:29–41.PubMedCrossRef
44.
go back to reference Morrissey MA, Kern N, Vale RD. CD47 ligation repositions the inhibitory receptor SIRPA to suppress integrin activation and Phagocytosis. Immunity. 2020;53:290–302e6.PubMedPubMedCentralCrossRef Morrissey MA, Kern N, Vale RD. CD47 ligation repositions the inhibitory receptor SIRPA to suppress integrin activation and Phagocytosis. Immunity. 2020;53:290–302e6.PubMedPubMedCentralCrossRef
45.
go back to reference Willingham SB, et al. The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors. Proc Natl Acad Sci U S A. 2012;109:6662–7.ADSPubMedPubMedCentralCrossRef Willingham SB, et al. The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors. Proc Natl Acad Sci U S A. 2012;109:6662–7.ADSPubMedPubMedCentralCrossRef
48.
go back to reference Zhang M, et al. Anti-CD47 treatment stimulates phagocytosis of Glioblastoma by M1 and M2 polarized macrophages and promotes M1 polarized macrophages in vivo. PLoS ONE. 2016;11:e0153550.PubMedPubMedCentralCrossRef Zhang M, et al. Anti-CD47 treatment stimulates phagocytosis of Glioblastoma by M1 and M2 polarized macrophages and promotes M1 polarized macrophages in vivo. PLoS ONE. 2016;11:e0153550.PubMedPubMedCentralCrossRef
49.
go back to reference Lecoultre M, Dutoit V, Walker PR. Phagocytic function of tumor-associated macrophages as a key determinant of tumor progression control: a review. J Immunother Cancer. 2020;8:e001408.PubMedPubMedCentralCrossRef Lecoultre M, Dutoit V, Walker PR. Phagocytic function of tumor-associated macrophages as a key determinant of tumor progression control: a review. J Immunother Cancer. 2020;8:e001408.PubMedPubMedCentralCrossRef
50.
go back to reference Dheilly E, et al. Selective blockade of the ubiquitous checkpoint receptor CD47 is enabled by dual-targeting bispecific antibodies. Mol Ther. 2017;25:523–33.PubMedPubMedCentralCrossRef Dheilly E, et al. Selective blockade of the ubiquitous checkpoint receptor CD47 is enabled by dual-targeting bispecific antibodies. Mol Ther. 2017;25:523–33.PubMedPubMedCentralCrossRef
51.
go back to reference Nath PR, et al. Natural killer cell recruitment and activation are regulated by CD47 expression in the tumor microenvironment. Cancer Immunol Res. 2019;7:1547–61.PubMedPubMedCentralCrossRef Nath PR, et al. Natural killer cell recruitment and activation are regulated by CD47 expression in the tumor microenvironment. Cancer Immunol Res. 2019;7:1547–61.PubMedPubMedCentralCrossRef
52.
go back to reference Anderson KL, et al. Evolutionarily conserved resistance to phagocytosis observed in melanoma cells is insensitive to upregulation of pro-phagocytic signals and to CD47 blockade. Melanoma Res. 2020;30:147–58.ADSPubMedCrossRef Anderson KL, et al. Evolutionarily conserved resistance to phagocytosis observed in melanoma cells is insensitive to upregulation of pro-phagocytic signals and to CD47 blockade. Melanoma Res. 2020;30:147–58.ADSPubMedCrossRef
53.
go back to reference Ngo M, et al. Antibody therapy targeting CD47 and CD271 effectively suppresses Melanoma Metastasis in patient-derived xenografts. Cell Rep. 2016;16:1701–16.PubMedCrossRef Ngo M, et al. Antibody therapy targeting CD47 and CD271 effectively suppresses Melanoma Metastasis in patient-derived xenografts. Cell Rep. 2016;16:1701–16.PubMedCrossRef
55.
56.
go back to reference Lu J, et al. Reprogramming of TAMs via the STAT3/CD47-SIRPα axis promotes acquired resistance to EGFR-TKIs in lung cancer. Cancer Lett. 2023;564:216205.PubMedCrossRef Lu J, et al. Reprogramming of TAMs via the STAT3/CD47-SIRPα axis promotes acquired resistance to EGFR-TKIs in lung cancer. Cancer Lett. 2023;564:216205.PubMedCrossRef
57.
go back to reference Moreno-Gonzalo O, et al. HDAC6 controls innate immune and autophagy responses to TLR-mediated signalling by the intracellular bacteria listeria monocytogenes. PLOS Pathog. 2017;13:e1006799.PubMedPubMedCentralCrossRef Moreno-Gonzalo O, et al. HDAC6 controls innate immune and autophagy responses to TLR-mediated signalling by the intracellular bacteria listeria monocytogenes. PLOS Pathog. 2017;13:e1006799.PubMedPubMedCentralCrossRef
58.
go back to reference Gauttier V et al. Selective SIRPα blockade reverses tumor T cell exclusion and overcomes cancer immunotherapy resistance. J Clin Invest 130, 6109–23. Gauttier V et al. Selective SIRPα blockade reverses tumor T cell exclusion and overcomes cancer immunotherapy resistance. J Clin Invest 130, 6109–23.
60.
go back to reference Schwartz AL, et al. Antisense targeting of CD47 enhances human cytotoxic T-cell activity and increases survival of mice bearing B16 melanoma when combined with anti-CTLA4 and tumor irradiation. Cancer Immunol Immunother. 2019;68:1805–17.PubMedPubMedCentralCrossRef Schwartz AL, et al. Antisense targeting of CD47 enhances human cytotoxic T-cell activity and increases survival of mice bearing B16 melanoma when combined with anti-CTLA4 and tumor irradiation. Cancer Immunol Immunother. 2019;68:1805–17.PubMedPubMedCentralCrossRef
61.
go back to reference Soto-Pantoja DR, et al. CD47 in the tumor microenvironment limits cooperation between antitumor T-cell immunity and radiotherapy. Cancer Res. 2014;74:6771–83.PubMedPubMedCentralCrossRef Soto-Pantoja DR, et al. CD47 in the tumor microenvironment limits cooperation between antitumor T-cell immunity and radiotherapy. Cancer Res. 2014;74:6771–83.PubMedPubMedCentralCrossRef
Metadata
Title
Targeting HDAC6 improves anti-CD47 immunotherapy
Authors
Maria Gracia-Hernandez
Ashutosh S. Yende
Nithya Gajendran
Zubaydah Alahmadi
Xintang Li
Zuleima Munoz
Karen Tan
Satish Noonepalle
Maho Shibata
Alejandro Villagra
Publication date
01-12-2024
Publisher
BioMed Central
Keywords
Melanoma
Melanoma
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2024
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-024-02982-4

Other articles of this Issue 1/2024

Journal of Experimental & Clinical Cancer Research 1/2024 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine