Chidamide plus Tyrosine Kinase Inhibitor Remodel the Tumor Immune Microenvironment and Reduce Tumor Progression When Combined with Immune Checkpoint Inhibitor in Naïve and Anti-PD-1 Resistant CT26-Bearing Mice
<p>(<b>A</b>–<b>F</b>) shows the results of therapeutic responses to Cabozantinib or Regorafenib combined with Chidamide-k30 plus anti-PD-1 antibody in CT26 tumor-bearing mice. Balb/c mice bearing a CT26 tumor were treated with various therapeutic modalities as indicated. The combination with Cabozantinib/ Regorafenib is shown in (<b>A</b>) consecutive treatment schedule (<b>B</b>) total tumor volumes (<b>C</b>) Endpoint evaluated tumor volumes at D25 (<b>D</b>) Mice body weights (<b>E</b>) Individual tumor volumes (<b>F</b>) animal survival rates. CT26 tumor-bearing mice were treated as indicated and euthanized at a tumor volume of 3000 mm<sup>3</sup> after tumor implantation. After 16-day treatment followed by tumor assessment, the mice with CR and PR response were re-inoculated with CT-26 cancer cells into the opposite flank in a rechallenge experiment. Red line squares indicates re-inoculated CT26 tumor growth in rechallenge experiment. Data are given as mean ± SD; × <span class="html-italic">p</span> < 0.05, ××× <span class="html-italic">p</span> < 0.001, ### <span class="html-italic">p</span> < 0.001, one-way ANOVA with Tukey’s test. ×, compared to IgG; #, compared to PD-1, & compared to PD-1 + Reg. (<span class="html-italic">n</span> = 6–8).</p> "> Figure 2
<p>Immunosuppressive cells in microenvironment were attenuated by anti-PD-1 +TKI +CD-k30 treatment. (<b>C</b>–<b>J</b>) show the results of immune cell population analysis of lymphocytes and myeloid-derived MDSCs in the CT26-bearing mice tumors. The CT26 tumor-bearing mice were treated with various therapeutic modalities as indicated. Tumor samples were isolated on day 20 after 9-day treatment for analyzing immune cell population in tumors. (<b>A</b>) Consecutive treatment schedule and (<b>B</b>) Tumor sizes of each treatment group. (<b>C</b>–<b>F</b>) show the results of flow cytometry of CD3, CD4, CD8, and Treg cell population in tumors. (<b>G</b>–<b>J</b>) show the results of flow cytometry of myeloid-derived CD11b, PMN-MDSC, M-MDSC, and tumor association macrophage (TAM) cell populations in tumors. Results are shown as mean ± SD; × <span class="html-italic">p</span> < 0.05, ×× <span class="html-italic">p</span> < 0.01, ××× <span class="html-italic">p</span> < 0.001, one-way ANOVA with Tukey’s test. ×, compared to IgG; #, compared to PD-1 (<span class="html-italic">n</span> = 6–7).</p> "> Figure 3
<p>Identification of target genes of anti-PD-1 Ab combined with Regorafenib/Cabozantinib plus Chidamide-k30 treatment, which significantly regulates gene expression in TME of CT26 tumors-bearing mice. Tumors were analyzed on day 20 after 9-day treatment for gene expression by RNA-seq. (<b>A</b>) Volcano plot of differentially expressed genes obtained by RNA-seq analysis in anti-PD-1 Ab combined with Regorafenib/Cabozantinib plus Chidamide-k30 treatment CT26 tumors compared to IgG control tumors. Significantly upregulated or downregulated genes are plotted in red and blue points, respectively. (<b>B</b>) Meta-enrichment analysis summary for significantly upregulated and downregulated genes was indicated by display of categories of related pathways and the number of affected genes of the corresponding pathway. The pathways highlighted with red color were related to the gene expression signatures in (<b>C</b>–<b>G</b>). (<b>C</b>–<b>G</b>) show that gene expression related to chemokine activity, cytokine activity, leukocyte migration, cell chemotaxis, T-cell-mediated cytotoxicity and angiogenesis were analyzed in tumors. (<b>C</b>) GSEA enrichment analysis of tumors treated with anti-PD-1. (<b>D</b>) GSEA enrichment analysis of tumors treated with PD-1 + cab. <b>(</b><b>E</b>) GSEA enrichment analysis of tumors treated with anti-PD-1 + Cab + CD-k30. (F) GSEA enrichment analysis of tumors treated with anti-PD-1 + Reg. (<b>G</b>) GSEA enrichment analysis of tumors treated with anti-PD-1 + Reg + CD-k30. NES: normalized enrichment score; FDR: false discovery rates. Signature scores were calculated by mean log2 (TPM) of their respective member genes; <span class="html-italic">p</span>-values: Mann-Whitney test, two-tailed. When <span class="html-italic">p</span> ≧ 0.05, the GSEA analysis panel(s) is outlined with a red dotted line. When gene expression was downregulated, the GSEA analysis panel(s) is outlined with a blue solid line.</p> "> Figure 4
<p>(<b>A</b>–<b>F</b>) shows the results of therapeutic responses and immunity evaluations of different ICIs combined with Cabozantinib or Regorafenib plus Chidamide-k30 in CT26 tumor-bearing mice. Balb/c mice bearing a CT26 tumor were treated with various therapeutic modalities as indicated. The combination therapy is shown in (<b>A</b>) consecutive treatment schedule and (<b>B</b>) total tumor volumes. (<b>C</b>) Endpoint evaluated tumor volumes at D26. (<b>D</b>) Mice body weights. (<b>E</b>) Individual tumor volumes. (<b>F</b>) Animal survival rates. CT26 tumor-bearing mice were treated as indicated and euthanized at a tumor volume of 3000 mm<sup>3</sup> after tumor implantation. After 16-day treatment followed by tumor assessment, the mice with CR and PR response were re-inoculated with CT-26 cancer cells into the opposite flank in a rechallenge experiment. Red line squares indicates re-inoculated CT26 tumor growth in rechallenge experiment. Data are given as mean ± SD; × <span class="html-italic">p</span> < 0.05, ××× <span class="html-italic">p</span> < 0.001 compared to IgG; #, compared to PD-1; one-way ANOVA with Tukey’s test. (<span class="html-italic">n</span> = 7–10).</p> "> Figure 5
<p>(<b>A</b>) shows the consecutive treatment schedule and responsive results of the first-line anti-PD-1 Ab treatment. Male Balb/c mice bearing subcutaneous CT26 tumors (1 × 10<sup>6</sup> cell/mice) were treated with a first-line therapy of anti-PD-1 Ab (mean tumor volume: 150 mm<sup>3</sup> when treatment began). The mice were intraperitoneally (i.p.) administered anti-PD-1 Ab or IgG at 2.5 mg/kg, once every 3 days for 3 doses. When the mice responded to anti-PD-1 Ab with tumor shrinking, they were given three more doses. (<b>B</b>) Tumor size (mm<sup>3</sup>) from mice responsive to first-line anti-PD-1 Ab treatment, in comparison with control group treated with anti-IgG antibody 3 times. *, <span class="html-italic">p</span> < 0.05. <a href="#ijms-23-10677-f005" class="html-fig">Figure 5</a>C–E show the results of a second-line treatment in the mice having anti-PD-1 antibody primary resistance. In the first-line anti-PD-1 antibody therapy, if the tumor showed 2.5- to 3-times consecutive increases in tumor volume and with volumes of <600 mm<sup>3</sup>, the mice were defined as having primary resistance. These mice were subsequently reenrolled and divided into nine groups in a second-line treatment for efficacy study. In the second-line treatment, anti-IgG antibody was as a Ab control, anti-PD-1, and anti-CTLA-4 antibodies were administered intraperitoneally (i.p.) at 2.5 mg/kg, once every 3 days for 6 doses. The combinations in the second-line treatment were: Chidamide-HCL salt (50 mg/kg) + Celecoxib (50 mg/kg) (CC-02), anti-PD-1(2.5 mg/kg) + CC-02, anti-CTLA-4 antibody (2.5 mg/kg) + CC-02 as shown in (<b>C</b>) Tumor size (mm<sup>3</sup>) Anti--CTLA-4 antibody (2.5 mg/kg) combined with Regorafenib (30 mg/kg) (reg) plus Chidamide-k30 (50 mg/kg) (CD-k30); and anti-CTLA-4 antibody (2.5 mg/kg) combined with Cabozantinib (30 mg/kg) (cab) plus Chidamide-k30 (50 mg/kg) (CD-k30) as shown in (<b>D</b>) tumor size (mm<sup>3</sup>). <a href="#ijms-23-10677-f001" class="html-fig">Figure 1</a>D also shows the results of second-line treatment in mice with hyperprogressive disease (HPD) tumor during anti-PD-1 antibody therapy. After three times of administration of first-line Anti-PD-1 antibody, if the tumor volumes were >600 mm<sup>3</sup>, the mice were defined as having hyperprogressive disease (HPD). These mice were subsequently reenrolled in a second-line treatment for efficacy study. The combination in the second-line treatment is Anti-CTLA-4 antibody (2.5 mg/kg) combined with Cabozantinib (30 mg/kg) plus Chidamide-HCl salt (50 mg/kg). (<b>E</b>) Efficacy evaluated endpoint tumor size (mm<sup>3</sup>) at D28, (<b>F</b>) Mice body weights. (<b>G</b>) show the results of individual tumor volume in the second-line treatment of the anti-PD-1 Ab primary resistance mice. (<b>H</b>,<b>I</b>) Overall survival rates after second-line treatment for mice with primary resistance to anti-PD-1 antibody. After 16-day treatment followed by tumor assessment, the mice with CR and PR response were re-inoculated with CT-26 cancer cells into the opposite flank in a rechallenge experiment. Red line squares indicates re-inoculated CT26 tumor growth in a rechallenge experiment. Data are given as mean ± SD; × <span class="html-italic">p</span> < 0.05, ×× <span class="html-italic">p</span> < 0.01, ××× <span class="html-italic">p</span> < 0.001 compared to IgG; #, compared to PD-1; &, compared to CTLA-4, one-way ANOVA with Tukey’s test. (<span class="html-italic">n</span> = 5–11).</p> "> Figure 6
<p>Identification of target genes of second-line treatment with anti-CTLA-4 Ab combined with CC-02 or Regorafenib/Cabozantinib plus Chidamide-k30 that significantly regulates gene expression in TME of CT26 tumor-bearing mice. Tumors were analyzed on day 12 after starting second-line treatment for gene expression by RNA-seq. (<b>A</b>) Volcano plot of differentially expressed genes obtained by RNA-seq analysis in treated CT26 tumors compared to IgG control tumors. Significantly upregulated or downregulated genes are plotted in red and blue points, respectively. (<b>B</b>) Meta-enrichment analysis summary for significantly upregulated and downregulated genes was indicated by display of categories of related pathways and the number of affected genes of the corresponding pathway. The pathways highlighted with red color were related to the gene expression signatures in (<b>C</b>–<b>G</b>). (<b>C</b>–<b>G</b>) show results of gene expression related to response to INF-γ, cellular response to INF-γ, leukocyte migration, cell chemotaxis, adaptive immune response, and angiogenesis being analyzed. (<b>C</b>) GSEA enrichment analysis of tumors treated with anti-CTLA-4. (D) GSEA enrichment analysis of tumors treated with CC-02. (<b>E</b>) GSEA enrichment analysis of tumors treated with anti-CTLA-4 + CC-02. (<b>F</b>) GSEA enrichment analysis of tumors treated with anti-CTLA-4 + Cab + CD-k30. (<b>G</b>) GSEA enrichment analysis of tumors treated with anti-CTLA-4 + Reg + CD-k30. NES: normalized enrichment score; FDR: false discovery rates. Signature scores were calculated by mean log2 (TPM) of their respective member genes; <span class="html-italic">p</span>-values: Mann-Whitney test, two-tailed. When <span class="html-italic">p</span> ≧ 0.05, the GSEA analysis panel(s) is outlined with a red dotted line. When gene expression was downregulated, the GSEA analysis panel(s) is outlined with a blue solid line.</p> ">
Abstract
:1. Introduction
2. Results
2.1. Anticancer Effect of VEGFR-TKIs Combined with Anti-PD-1 Ab in CT26-Bearing Mice
2.2. Anticancer Activity of Anti-PD-1 Antibody Combined with Cabozantinib or Regorafenib Plus Chidamide-k30 in CT26-Bearing Mice
2.3. Chidamide Is a Key Component in Triplet Combination Regimens for Significantly Regulating Immune Cell Population and Gene Expression in the TME of CT26 Tumor-Bearing Mice
2.4. Anticancer Activity of Several ICIs Combined with Cabozantinib/Regorafenib Plus Chidamide in CT26-Bearing Mice
2.5. Overcoming First-Line Anti-PD-1 Antibody Treatment-Induced Drug Resistance Using Cabozantinib/Regorafenib plus Chidamide Combined with Anti-CTLA-4 Antibody in CT26-Bearing Mice
2.6. IntraTumor Gene Expression in First-Line Anti-PD-1 Antibody-Resistant CT-26 Tumor-Bearing Mice after Second-Line Therapy with Chidamide-k30 Combined with Cabozantinib/Regorafenib Plus Anti-CTLA-4 Antibody
3. Discussions
4. Material and Methods
4.1. Anti-Colorectal Cancer Activity in Animal Models
4.2. Tumor Rechallenge in Animal Models
4.3. Survival Rate in Animal Models
4.4. Flow Cytometry
4.5. Overcoming Primary Resistance and HPD Induced by First-Line PD-1 Checkpoint Blockade Therapy
4.6. RNA Quantification and Qualification
4.7. Library Preparation for Transcriptome Sequencing
4.8. Bioinformatics
4.9. Statistics
5. Conclusions
Supplementary Materials
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Liu, D. CAR-T “the living drugs”, immune checkpoint inhibitors, and precision medicine: A new era of cancer therapy. J. Hematol. Oncol. 2019, 12, 113. [Google Scholar] [CrossRef] [PubMed]
- Chyuan, I.T.; Chu, C.L.; Hsu, P.N. Targeting the Tumor Microenvironment for Improving Therapeutic Effectiveness in Cancer Immunotherapy: Focusing on Immune Checkpoint Inhibitors and Combination Therapies. Cancers 2021, 13, 1188. [Google Scholar] [CrossRef] [PubMed]
- Robert, C. A decade of immune-checkpoint inhibitors in cancer therapy. Nat. Commun. 2020, 11, 3801. [Google Scholar] [CrossRef]
- Twomey, J.D.; Zhang, B. Cancer Immunotherapy Update: FDA-Approved Checkpoint Inhibitors and Companion Diagnostics. AAPS J. 2021, 23, 39. [Google Scholar] [CrossRef]
- Robert, C.; Ribas, A.; Hamid, O.; Daud, A.; Wolchok, J.D.; Joshua, A.M.; Hwu, W.J.; Weber, J.S.; Gangadhar, T.C.; Joseph, R.W.; et al. Durable Complete Response After Discontinuation of Pembrolizumab in Patients With Metastatic Melanoma. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2018, 36, 1668–1674. [Google Scholar] [CrossRef]
- Adam, T.; Becker, T.M.; Chua, W.; Bray, V.; Roberts, T.L. The Multiple Potential Biomarkers for Predicting Immunotherapy Response-Finding the Needle in the Haystack. Cancers 2021, 13, 277. [Google Scholar] [CrossRef] [PubMed]
- Jenkins, R.W.; Barbie, D.A.; Flaherty, K.T. Mechanisms of resistance to immune checkpoint inhibitors. Br. J. Cancer 2018, 118, 9–16. [Google Scholar] [CrossRef]
- Bagchi, S.; Yuan, R.; Engleman, E.G. Immune Checkpoint Inhibitors for the Treatment of Cancer: Clinical Impact and Mechanisms of Response and Resistance. Annu. Rev. Pathol. 2021, 16, 223–249. [Google Scholar] [CrossRef]
- Kamada, T.; Togashi, Y.; Tay, C.; Ha, D.; Sasaki, A.; Nakamura, Y.; Sato, E.; Fukuoka, S.; Tada, Y.; Tanaka, A.; et al. PD-1(+) regulatory T cells amplified by PD-1 blockade promote hyperprogression of cancer. Proc. Natl. Acad. Sci. USA 2019, 116, 9999–10008. [Google Scholar] [CrossRef]
- Vukadin, S.; Khaznadar, F.; Kizivat, T.; Vcev, A.; Smolic, M. Molecular Mechanisms of Resistance to Immune Checkpoint Inhibitors in Melanoma Treatment: An Update. Biomedicines 2021, 9, 835. [Google Scholar] [CrossRef]
- Wang, S.; He, Z.; Wang, X.; Li, H.; Liu, X.S. Antigen presentation and tumor immunogenicity in cancer immunotherapy response prediction. eLife 2019, 8, e49020. [Google Scholar] [CrossRef] [PubMed]
- Yang, J.; Yan, J.; Liu, B. Targeting VEGF/VEGFR to Modulate Antitumor Immunity. Front. Immunol. 2018, 9, 978. [Google Scholar] [CrossRef] [PubMed]
- Narita, Y.; Kitamura, H.; Wakita, D.; Sumida, K.; Masuko, K.; Terada, S.; Nakano, K.; Nishimura, T. The key role of IL-6-arginase cascade for inducing dendritic cell-dependent CD4(+) T cell dysfunction in tumor-bearing mice. J. Immunol. 2013, 190, 812–820. [Google Scholar] [CrossRef] [PubMed]
- Ruffell, B.; Chang-Strachan, D.; Chan, V.; Rosenbusch, A.; Ho, C.M.; Pryer, N.; Daniel, D.; Hwang, E.S.; Rugo, H.S.; Coussens, L.M. Macrophage IL-10 blocks CD8+ T cell-dependent responses to chemotherapy by suppressing IL-12 expression in intratumoral dendritic cells. Cancer Cell 2014, 26, 623–637. [Google Scholar] [CrossRef]
- Dhatchinamoorthy, K.; Colbert, J.D.; Rock, K.L. Cancer Immune Evasion Through Loss of MHC Class I Antigen Presentation. Front. Immunol. 2021, 12, 636568. [Google Scholar] [CrossRef] [PubMed]
- Lee, Y.J.; Lee, J.B.; Ha, S.J.; Kim, H.R. Clinical Perspectives to Overcome Acquired Resistance to Anti-Programmed Death-1 and Anti-Programmed Death Ligand-1 Therapy in Non-Small Cell Lung Cancer. Mol. Cells 2021, 44, 363–373. [Google Scholar] [CrossRef]
- Wang, X.; Wang, F.; Zhong, M.; Yarden, Y.; Fu, L. The biomarkers of hyperprogressive disease in PD-1/PD-L1 blockage therapy. Mol. Cancer 2020, 19, 81. [Google Scholar] [CrossRef]
- Khan, K.A.; Kerbel, R.S. Improving immunotherapy outcomes with anti-angiogenic treatments and vice versa. Nat. Rev. Clin. Oncol. 2018, 15, 310–324. [Google Scholar] [CrossRef]
- Tada, Y.; Togashi, Y.; Kotani, D.; Kuwata, T.; Sato, E.; Kawazoe, A.; Doi, T.; Wada, H.; Nishikawa, H.; Shitara, K. Targeting VEGFR2 with Ramucirumab strongly impacts effector/ activated regulatory T cells and CD8(+) T cells in the tumor microenvironment. J. Immunother. Cancer 2018, 6, 106. [Google Scholar] [CrossRef]
- Meder, L.; Schuldt, P.; Thelen, M.; Schmitt, A.; Dietlein, F.; Klein, S.; Borchmann, S.; Wennhold, K.; Vlasic, I.; Oberbeck, S.; et al. Combined VEGF and PD-L1 Blockade Displays Synergistic Treatment Effects in an Autochthonous Mouse Model of Small Cell Lung Cancer. Cancer Res. 2018, 78, 4270–4281. [Google Scholar] [CrossRef] [Green Version]
- Wang, Q.; Gao, J.; Di, W.; Wu, X. Anti-angiogenesis therapy overcomes the innate resistance to PD-1/PD-L1 blockade in VEGFA-overexpressed mouse tumor models. Cancer Immunol. Immunother. CII 2020, 69, 1781–1799. [Google Scholar] [CrossRef] [PubMed]
- Jung, K.; Heishi, T.; Khan, O.F.; Kowalski, P.S.; Incio, J.; Rahbari, N.N.; Chung, E.; Clark, J.W.; Willett, C.G.; Luster, A.D.; et al. Ly6Clo monocytes drive immunosuppression and confer resistance to anti-VEGFR2 cancer therapy. J. Clin. Investig. 2017, 127, 3039–3051. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.D.; Hoang, A.; Zhou, L.; Kalra, S.; Yetil, A.; Sun, M.; Ding, Z.; Zhang, X.; Bai, S.; German, P.; et al. Resistance to Antiangiogenic Therapy Is Associated with an Immunosuppressive Tumor Microenvironment in Metastatic Renal Cell Carcinoma. Cancer Immunol. Res. 2015, 3, 1017–1029. [Google Scholar] [CrossRef] [PubMed]
- Noman, M.Z.; Desantis, G.; Janji, B.; Hasmim, M.; Karray, S.; Dessen, P.; Bronte, V.; Chouaib, S. PD-L1 is a novel direct target of HIF-1alpha, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J. Exp. Med. 2014, 211, 781–790. [Google Scholar] [CrossRef] [PubMed]
- Sukumar, M.; Liu, J.; Ji, Y.; Subramanian, M.; Crompton, J.G.; Yu, Z.; Roychoudhuri, R.; Palmer, D.C.; Muranski, P.; Karoly, E.D.; et al. Inhibiting glycolytic metabolism enhances CD8+ T cell memory and antitumor function. J. Clin. Investig. 2013, 123, 4479–4488. [Google Scholar] [CrossRef] [PubMed]
- Kheshtchin, N.; Arab, S.; Ajami, M.; Mirzaei, R.; Ashourpour, M.; Mousavi, N.; Khosravianfar, N.; Jadidi-Niaragh, F.; Namdar, A.; Noorbakhsh, F.; et al. Inhibition of HIF-1alpha enhances anti-tumor effects of dendritic cell-based vaccination in a mouse model of breast cancer. Cancer Immunol. Immunother. CII 2016, 65, 1159–1167. [Google Scholar] [CrossRef]
- Qian, D.Z.; Kachhap, S.K.; Collis, S.J.; Verheul, H.M.; Carducci, M.A.; Atadja, P.; Pili, R. Class II histone deacetylases are associated with VHL-independent regulation of hypoxia-inducible factor 1 alpha. Cancer Res. 2006, 66, 8814–8821. [Google Scholar] [CrossRef]
- Miyake, K.; Yoshizumi, T.; Imura, S.; Sugimoto, K.; Batmunkh, E.; Kanemura, H.; Morine, Y.; Shimada, M. Expression of hypoxia-inducible factor-1alpha, histone deacetylase 1, and metastasis-associated protein 1 in pancreatic carcinoma: Correlation with poor prognosis with possible regulation. Pancreas 2008, 36, e1–e9. [Google Scholar] [CrossRef]
- Chen, J.S.; Chou, C.H.; Wu, Y.H.; Yang, M.H.; Chu, S.H.; Chao, Y.S.; Chen, C.N. CC-01 (chidamide plus celecoxib) modifies the tumor immune microenvironment and reduces tumor progression combined with immune checkpoint inhibitor. Sci. Rep. 2022, 12, 1100. [Google Scholar] [CrossRef]
- Jorgovanovic, D.; Song, M.; Wang, L.; Zhang, Y. Roles of IFN-gamma in tumor progression and regression: A review. Biomark. Res. 2020, 8, 49. [Google Scholar] [CrossRef]
- Bekaii-Saab, T.; Kim, R.; Kim, T.W.; O’Connor, J.M.; Strickler, J.H.; Malka, D.; Sartore-Bianchi, A.; Bi, F.; Yamaguchi, K.; Yoshino, T.; et al. Third- or Later-line Therapy for Metastatic Colorectal Cancer: Reviewing Best Practice. Clin. Colorectal Cancer 2019, 18, e117–e129. [Google Scholar] [CrossRef] [PubMed]
- Wilhelm, S.M.; Dumas, J.; Adnane, L.; Lynch, M.; Carter, C.A.; Schutz, G.; Thierauch, K.H.; Zopf, D. Regorafenib (BAY 73-4506): A new oral multikinase inhibitor of angiogenic, stromal and oncogenic receptor tyrosine kinases with potent preclinical antitumor activity. Int. J. Cancer 2011, 129, 245–255. [Google Scholar] [CrossRef] [PubMed]
- Mukherjee, S.K.; Wilhelm, A.; Antoniades, C.G. TAM receptor tyrosine kinase function and the immunopathology of liver disease. Am. J. Physiol. Gastrointest. Liver Physiol. 2016, 310, G899–G905. [Google Scholar] [CrossRef]
- Bruix, J.; Qin, S.; Merle, P.; Granito, A.; Huang, Y.H.; Bodoky, G.; Pracht, M.; Yokosuka, O.; Rosmorduc, O.; Breder, V.; et al. Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017, 389, 56–66. [Google Scholar] [CrossRef]
- Abou-Alfa, G.K.; Meyer, T.; Cheng, A.L.; El-Khoueiry, A.B.; Rimassa, L.; Ryoo, B.Y.; Cicin, I.; Merle, P.; Chen, Y.; Park, J.W.; et al. Cabozantinib in Patients with Advanced and Progressing Hepatocellular Carcinoma. N. Engl. J. Med. 2018, 379, 54–63. [Google Scholar] [CrossRef]
- Efremova, M.; Rieder, D.; Klepsch, V.; Charoentong, P.; Finotello, F.; Hackl, H.; Hermann-Kleiter, N.; Lower, M.; Baier, G.; Krogsdam, A.; et al. Targeting immune checkpoints potentiates immunoediting and changes the dynamics of tumor evolution. Nat. Commun. 2018, 9, 32. [Google Scholar] [CrossRef]
- Zhong, W.; Myers, J.S.; Wang, F.; Wang, K.; Lucas, J.; Rosfjord, E.; Lucas, J.; Hooper, A.T.; Yang, S.; Lemon, L.A.; et al. Comparison of the molecular and cellular phenotypes of common mouse syngeneic models with human tumors. BMC Genom. 2020, 21, 2. [Google Scholar] [CrossRef]
- Le, D.T.; Durham, J.N.; Smith, K.N.; Wang, H.; Bartlett, B.R.; Aulakh, L.K.; Lu, S.; Kemberling, H.; Wilt, C.; Luber, B.S.; et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017, 357, 409–413. [Google Scholar] [CrossRef]
- Doleschel, D.; Hoff, S.; Koletnik, S.; Rix, A.; Zopf, D.; Kiessling, F.; Lederle, W. Regorafenib enhances anti-PD1 immunotherapy efficacy in murine colorectal cancers and their combination prevents tumor regrowth. J. Exp. Clin. Cancer Res. CR 2021, 40, 288. [Google Scholar] [CrossRef]
- Liu, L.; Mayes, P.A.; Eastman, S.; Shi, H.; Yadavilli, S.; Zhang, T.; Yang, J.; Seestaller-Wehr, L.; Zhang, S.Y.; Hopson, C.; et al. The BRAF and MEK Inhibitors Dabrafenib and Trametinib: Effects on Immune Function and in Combination with Immunomodulatory Antibodies Targeting PD-1, PD-L1, and CTLA-4. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2015, 21, 1639–1651. [Google Scholar] [CrossRef] [Green Version]
- Hansen, W.; Hutzler, M.; Abel, S.; Alter, C.; Stockmann, C.; Kliche, S.; Albert, J.; Sparwasser, T.; Sakaguchi, S.; Westendorf, A.M.; et al. Neuropilin 1 deficiency on CD4+Foxp3+ regulatory T cells impairs mouse melanoma growth. J. Exp. Med. 2012, 209, 2001–2016. [Google Scholar] [CrossRef] [PubMed]
- Cheng, A.L.; Hsu, C.; Chan, S.L.; Choo, S.P.; Kudo, M. Challenges of combination therapy with immune checkpoint inhibitors for hepatocellular carcinoma. J. Hepatol. 2020, 72, 307–319. [Google Scholar] [CrossRef] [PubMed]
- Shigeta, K.; Matsui, A.; Kikuchi, H.; Klein, S.; Mamessier, E.; Chen, I.X.; Aoki, S.; Kitahara, S.; Inoue, K.; Shigeta, A.; et al. Regorafenib combined with PD1 blockade increases CD8 T-cell infiltration by inducing CXCL10 expression in hepatocellular carcinoma. J. Immunother. Cancer 2020, 8, e001435. [Google Scholar] [CrossRef]
- Zopf, D.; Fichtner, I.; Bhargava, A.; Steinke, W.; Thierauch, K.H.; Diefenbach, K.; Wilhelm, S.; Hafner, F.T.; Gerisch, M. Pharmacologic activity and pharmacokinetics of metabolites of regorafenib in preclinical models. Cancer Med. 2016, 5, 3176–3185. [Google Scholar] [CrossRef] [PubMed]
- Terme, M.; Pernot, S.; Marcheteau, E.; Sandoval, F.; Benhamouda, N.; Colussi, O.; Dubreuil, O.; Carpentier, A.F.; Tartour, E.; Taieb, J. VEGFA-VEGFR pathway blockade inhibits tumor-induced regulatory T-cell proliferation in colorectal cancer. Cancer Res. 2013, 73, 539–549. [Google Scholar] [CrossRef] [PubMed]
- Motz, G.T.; Coukos, G. Deciphering and reversing tumor immune suppression. Immunity 2013, 39, 61–73. [Google Scholar] [CrossRef]
- Liu, C.; He, H.; Li, X.; Su, M.A.; Cao, Y. Correction: Dynamic metrics-based biomarkers to predict responders to anti-PD-1 immunotherapy. Br. J. Cancer 2019, 120, 772. [Google Scholar] [CrossRef]
- Lund, A.W.; Duraes, F.V.; Hirosue, S.; Raghavan, V.R.; Nembrini, C.; Thomas, S.N.; Issa, A.; Hugues, S.; Swartz, M.A. VEGF-C promotes immune tolerance in B16 melanomas and cross-presentation of tumor antigen by lymph node lymphatics. Cell Rep. 2012, 1, 191–199. [Google Scholar] [CrossRef]
- Peng, D.; Kryczek, I.; Nagarsheth, N.; Zhao, L.; Wei, S.; Wang, W.; Sun, Y.; Zhao, E.; Vatan, L.; Szeliga, W.; et al. Epigenetic silencing of TH1-type chemokines shapes tumour immunity and immunotherapy. Nature 2015, 527, 249–253. [Google Scholar] [CrossRef]
- Omar, N.E.; El-Fass, K.A.; Abushouk, A.I.; Elbaghdady, N.; Barakat, A.E.M.; Noreldin, A.E.; Johar, D.; Yassin, M.; Hamad, A.; Elazzazy, S.; et al. Diagnosis and Management of Hematological Adverse Events Induced by Immune Checkpoint Inhibitors: A Systematic Review. Front. Immunol. 2020, 11, 1354. [Google Scholar] [CrossRef]
- Schwartz, G.; Darling, J.O.; Mindo, M.; Damicis, L. Management of Adverse Events Associated with Cabozantinib Treatment in Patients with Advanced Hepatocellular Carcinoma. Target. Oncol. 2020, 15, 549–565. [Google Scholar] [CrossRef] [PubMed]
- Jiang, Z.; Li, W.; Hu, X.; Zhang, Q.; Sun, T.; Cui, S.; Wang, S.; Ouyang, Q.; Yin, Y.; Geng, C.; et al. Tucidinostat plus exemestane for postmenopausal patients with advanced, hormone receptor-positive breast cancer (ACE): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. Oncol. 2019, 20, 806–815. [Google Scholar] [CrossRef]
Regimens | Initial Tumor Volume (mm3) | ORR (%) | PD | SD | PR | CR | Survival Rate (%) | Relapse * (Recurrence) (%) | Immunity # (Rechallenge) (%) |
---|---|---|---|---|---|---|---|---|---|
anti-PD-1 Ab | 227 | 0 | 8 | 0 | 0 | 0 | 0 | - | - |
anti-PD-1 Ab + reg | 13 | 1 | 6 | 0 | 1 | 13 | 0 | 100 | |
anti-PD-1 Ab + cab | 0 | 1 | 5 | 0 | 0 | 0 | - | - | |
anti-PD-1 Ab + cab+ CD-k30 | 50 | 0 | 4 | 1 | 3 | 50 | 50 | 100 | |
anti-PD-1 Ab + reg + CD-k30 | 43 | 0 | 4 | 0 | 3 | 86 | 0 | 100 |
Regimens | Initial Tumor Volume (mm3) | ORR (%) | PD | SD | PR | CR | ORR (%) & | PD & | SD & | PR & | CR & | Survival Rate (%) | Relapse * (Recurrence) (%) | Immunity # (Rechallenge) (%) |
---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
anti-PD-1 Ab | 243 | 0 | 6 | 4 | 0 | 0 | 0 | 8 | 2 | 0 | 0 | 0 | - | - |
anti-PD-1 Ab + reg + CD-k30 | 30 | 0 | 7 | 1 | 2 | 60 | 1 | 3 | 2 | 4 | 80 | 0 | 100 | |
anti-PD-L1 Ab + reg + CD-k30 | 89 | 0 | 1 | 3 | 5 | 89 | 0 | 1 | 0 | 8 | 89 | 13 | 100 | |
anti-CTLA-4 Ab + reg + CD-k30 | 60 | 0 | 4 | 2 | 4 | 80 | 1 | 1 | 1 | 7 | 90 | 0 | 100 | |
anti-PD-1 Ab + cab+ CD-k30 | 40 | 1 | 5 | 0 | 4 | 60 | 3 | 1 | 1 | 5 | 70 | 0 | 100 | |
Anti-PD-L1 Ab + cab + CD-k30 | 60 | 0 | 4 | 3 | 3 | 60 | 2 | 2 | 1 | 5 | 70 | 17 | 100 | |
anti-CTLA-4 Ab + cab + CD-k30 | 90 | 0 | 1 | 1 | 8 | 90 | 0 | 1 | 0 | 9 | 100 | 0 | 100 |
Number of Mice | Response to First-Line Anti-PD-1 Antibody Therapy | Types of Drug Resistance to First-Line Anti-PD-1 Antibody Therapy |
---|---|---|
10 | Treatment with anti-IgG antibody (as negative control) | N/A |
17 | Yes | Response * |
91 | NO | Primary resistance ** |
11 | NO | HPD *** |
Therapeutic Resistance | Regimens | Initial Tumor Volume (mm3) | ORR (%) | PD | SD | PR | CR | ORR (%) & | PD & | SD & | PR & | CR & | Survival Rate (%) | Relapse * (%) | Immunity # (%) |
---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
Primary resistance | anti-PD-1 | 396 | 0 | 4 | 1 | 0 | 0 | 0 | 5 | 0 | 0 | 0 | 0 | - | - |
Anti-CTLA-4 | 0 | 0 | 7 | 0 | 0 | 0 | 2 | 5 | 0 | 0 | 0 | - | - | ||
Anti-CTLA-4 + CC-02 | 37.5 | 1 | 4 | 0 | 3 | 37.5 | 2 | 3 | 0 | 3 | 37.5 | 0 | 100 | ||
Anti-CTLA-4 + reg+ CD-k30 | 62.5 | 0 | 2 | 1 | 5 | 87.5 | 0 | 1 | 0 | 7 | 87.5 | 0 | 100 | ||
Anti-CTLA-4 + cab + CD-k30 | 57.1 | 0 | 3 | 1 | 3 | 100 | 0 | 0 | 3 | 4 | 71.4 | 0 | 100 | ||
HPD | Anti-CTLA-4 + cab + CDHCl | 669 | 18.1 | 0 | 9 | 0 | 2 | 45.4% | 3 | 3 | 3 | 2 | 45.4 | 0 | 100 |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Chen, J.-S.; Hsieh, Y.-C.; Chou, C.-H.; Wu, Y.-H.; Yang, M.-H.; Chu, S.-H.; Chao, Y.-S.; Chen, C.-N. Chidamide plus Tyrosine Kinase Inhibitor Remodel the Tumor Immune Microenvironment and Reduce Tumor Progression When Combined with Immune Checkpoint Inhibitor in Naïve and Anti-PD-1 Resistant CT26-Bearing Mice. Int. J. Mol. Sci. 2022, 23, 10677. https://doi.org/10.3390/ijms231810677
Chen J-S, Hsieh Y-C, Chou C-H, Wu Y-H, Yang M-H, Chu S-H, Chao Y-S, Chen C-N. Chidamide plus Tyrosine Kinase Inhibitor Remodel the Tumor Immune Microenvironment and Reduce Tumor Progression When Combined with Immune Checkpoint Inhibitor in Naïve and Anti-PD-1 Resistant CT26-Bearing Mice. International Journal of Molecular Sciences. 2022; 23(18):10677. https://doi.org/10.3390/ijms231810677
Chicago/Turabian StyleChen, Jia-Shiong, Yi-Chien Hsieh, Cheng-Han Chou, Yi-Hong Wu, Mu-Hsuan Yang, Sz-Hao Chu, Ye-Su Chao, and Chia-Nan Chen. 2022. "Chidamide plus Tyrosine Kinase Inhibitor Remodel the Tumor Immune Microenvironment and Reduce Tumor Progression When Combined with Immune Checkpoint Inhibitor in Naïve and Anti-PD-1 Resistant CT26-Bearing Mice" International Journal of Molecular Sciences 23, no. 18: 10677. https://doi.org/10.3390/ijms231810677
APA StyleChen, J. -S., Hsieh, Y. -C., Chou, C. -H., Wu, Y. -H., Yang, M. -H., Chu, S. -H., Chao, Y. -S., & Chen, C. -N. (2022). Chidamide plus Tyrosine Kinase Inhibitor Remodel the Tumor Immune Microenvironment and Reduce Tumor Progression When Combined with Immune Checkpoint Inhibitor in Naïve and Anti-PD-1 Resistant CT26-Bearing Mice. International Journal of Molecular Sciences, 23(18), 10677. https://doi.org/10.3390/ijms231810677