Anti-PD-1/PD-L1 therapy augments lenvatinib's efficacy by favorably altering the immune microenvironment of murine anaplastic thyroid cancer
Viswanath Gunda
Department of Surgery, Division of General and Gastrointestinal Surgery, Harvard Medical School, Massachusetts General Hospital, Wang Ambulatory Care Center, 460, 15 Parkman Street, Boston, MA
V.G. and B.G. contributed equally to this work.Search for more papers by this authorBenjamin Gigliotti
Department of Surgery, Division of General and Gastrointestinal Surgery, Harvard Medical School, Massachusetts General Hospital, Wang Ambulatory Care Center, 460, 15 Parkman Street, Boston, MA
V.G. and B.G. contributed equally to this work.Search for more papers by this authorTameem Ashry
Department of Surgery, Division of General and Gastrointestinal Surgery, Harvard Medical School, Massachusetts General Hospital, Wang Ambulatory Care Center, 460, 15 Parkman Street, Boston, MA
Search for more papers by this authorDorothy Ndishabandi
Department of Surgery, Division of General and Gastrointestinal Surgery, Harvard Medical School, Massachusetts General Hospital, Wang Ambulatory Care Center, 460, 15 Parkman Street, Boston, MA
Search for more papers by this authorMichael McCarthy
Department of Surgery, Division of General and Gastrointestinal Surgery, Harvard Medical School, Massachusetts General Hospital, Wang Ambulatory Care Center, 460, 15 Parkman Street, Boston, MA
Search for more papers by this authorZhiheng Zhou
Department of Surgery, Division of General and Gastrointestinal Surgery, Harvard Medical School, Massachusetts General Hospital, Wang Ambulatory Care Center, 460, 15 Parkman Street, Boston, MA
Search for more papers by this authorSalma Amin
Department of Surgery, Division of General and Gastrointestinal Surgery, Harvard Medical School, Massachusetts General Hospital, Wang Ambulatory Care Center, 460, 15 Parkman Street, Boston, MA
Search for more papers by this authorKyu Eun Lee
Department of Surgery, Division of General and Gastrointestinal Surgery, Harvard Medical School, Massachusetts General Hospital, Wang Ambulatory Care Center, 460, 15 Parkman Street, Boston, MA
Search for more papers by this authorTabea Stork
Department of Surgery, Division of General and Gastrointestinal Surgery, Harvard Medical School, Massachusetts General Hospital, Wang Ambulatory Care Center, 460, 15 Parkman Street, Boston, MA
Search for more papers by this authorLori Wirth
Department of Surgery, Division of General and Gastrointestinal Surgery, Harvard Medical School, Massachusetts General Hospital, Wang Ambulatory Care Center, 460, 15 Parkman Street, Boston, MA
Search for more papers by this authorGordon J. Freeman
Department of Surgery, Division of General and Gastrointestinal Surgery, Harvard Medical School, Massachusetts General Hospital, Wang Ambulatory Care Center, 460, 15 Parkman Street, Boston, MA
Search for more papers by this authorAlessandro Alessandrini
Department of Surgery, Division of General and Gastrointestinal Surgery, Harvard Medical School, Massachusetts General Hospital, Wang Ambulatory Care Center, 460, 15 Parkman Street, Boston, MA
Search for more papers by this authorCorresponding Author
Sareh Parangi
Department of Surgery, Division of General and Gastrointestinal Surgery, Harvard Medical School, Massachusetts General Hospital, Wang Ambulatory Care Center, 460, 15 Parkman Street, Boston, MA
Correspondence to: Sareh Parangi, MD, Harvard Medical School, Massachusetts General Hospital, Department of Surgery, Division of General and Gastrointestinal Surgery, Wang Ambulatory Care Center 460, 15 Parkman Street, Boston, MA 02115, Tel.: 617-643-4806; Fax: 617-643-4802; E-mail: [email protected]Search for more papers by this authorViswanath Gunda
Department of Surgery, Division of General and Gastrointestinal Surgery, Harvard Medical School, Massachusetts General Hospital, Wang Ambulatory Care Center, 460, 15 Parkman Street, Boston, MA
V.G. and B.G. contributed equally to this work.Search for more papers by this authorBenjamin Gigliotti
Department of Surgery, Division of General and Gastrointestinal Surgery, Harvard Medical School, Massachusetts General Hospital, Wang Ambulatory Care Center, 460, 15 Parkman Street, Boston, MA
V.G. and B.G. contributed equally to this work.Search for more papers by this authorTameem Ashry
Department of Surgery, Division of General and Gastrointestinal Surgery, Harvard Medical School, Massachusetts General Hospital, Wang Ambulatory Care Center, 460, 15 Parkman Street, Boston, MA
Search for more papers by this authorDorothy Ndishabandi
Department of Surgery, Division of General and Gastrointestinal Surgery, Harvard Medical School, Massachusetts General Hospital, Wang Ambulatory Care Center, 460, 15 Parkman Street, Boston, MA
Search for more papers by this authorMichael McCarthy
Department of Surgery, Division of General and Gastrointestinal Surgery, Harvard Medical School, Massachusetts General Hospital, Wang Ambulatory Care Center, 460, 15 Parkman Street, Boston, MA
Search for more papers by this authorZhiheng Zhou
Department of Surgery, Division of General and Gastrointestinal Surgery, Harvard Medical School, Massachusetts General Hospital, Wang Ambulatory Care Center, 460, 15 Parkman Street, Boston, MA
Search for more papers by this authorSalma Amin
Department of Surgery, Division of General and Gastrointestinal Surgery, Harvard Medical School, Massachusetts General Hospital, Wang Ambulatory Care Center, 460, 15 Parkman Street, Boston, MA
Search for more papers by this authorKyu Eun Lee
Department of Surgery, Division of General and Gastrointestinal Surgery, Harvard Medical School, Massachusetts General Hospital, Wang Ambulatory Care Center, 460, 15 Parkman Street, Boston, MA
Search for more papers by this authorTabea Stork
Department of Surgery, Division of General and Gastrointestinal Surgery, Harvard Medical School, Massachusetts General Hospital, Wang Ambulatory Care Center, 460, 15 Parkman Street, Boston, MA
Search for more papers by this authorLori Wirth
Department of Surgery, Division of General and Gastrointestinal Surgery, Harvard Medical School, Massachusetts General Hospital, Wang Ambulatory Care Center, 460, 15 Parkman Street, Boston, MA
Search for more papers by this authorGordon J. Freeman
Department of Surgery, Division of General and Gastrointestinal Surgery, Harvard Medical School, Massachusetts General Hospital, Wang Ambulatory Care Center, 460, 15 Parkman Street, Boston, MA
Search for more papers by this authorAlessandro Alessandrini
Department of Surgery, Division of General and Gastrointestinal Surgery, Harvard Medical School, Massachusetts General Hospital, Wang Ambulatory Care Center, 460, 15 Parkman Street, Boston, MA
Search for more papers by this authorCorresponding Author
Sareh Parangi
Department of Surgery, Division of General and Gastrointestinal Surgery, Harvard Medical School, Massachusetts General Hospital, Wang Ambulatory Care Center, 460, 15 Parkman Street, Boston, MA
Correspondence to: Sareh Parangi, MD, Harvard Medical School, Massachusetts General Hospital, Department of Surgery, Division of General and Gastrointestinal Surgery, Wang Ambulatory Care Center 460, 15 Parkman Street, Boston, MA 02115, Tel.: 617-643-4806; Fax: 617-643-4802; E-mail: [email protected]Search for more papers by this authorAbstract
Patients with anaplastic thyroid cancer (ATC) have an extremely poor prognosis despite multimodal therapy with surgery and chemoradiation. Lenvatinib, a multi-targeted tyrosine kinase inhibitor, as well as checkpoint inhibitors targeting the programmed cell death pathway, have proven effective in some patients with advanced thyroid cancer. Combination of these therapies is a potential means to boost effectiveness and minimize treatment resistance in ATC. We utilized our novel immunocompetent murine model of orthotopic ATC to demonstrate that lenvatinib led to significant tumor shrinkage and increased survival, while combination therapy led to dramatic improvements in both. Lenvatinib monotherapy increased tumor-infiltrating macrophages, CD8+ T-cells, regulatory T-cells, and most notably, polymorphonuclear myeloid derived suppressor cells (PMN-MDSCs). While both combination therapies led to further increases in CD8+ T-cells, only the lenvatinib and anti-PD-1 combination decreased PMN-MDSCs. PMN-MDSC expansion was also seen in the blood of mice and one patient receiving lenvatinib therapy for ATC. RNA-Seq of the ATC cell line used in our mouse model demonstrated that lenvatinib has multifaceted effects on angiogenesis, response to hypoxia, the epithelial-to-mesenchymal transition, and on multiple pathways implicated in inflammation and host immunity. Combination of lenvatinib with anti-Gr-1 antibody ameliorated lenvatinib's expansion of MDSCs and significantly improved lenvatinib's anti-tumor effect. These data suggest that MDSCs play a negative role in ATC's response to lenvatinib and support future study of their role as a potential biomarker and treatment target.
Abstract
What's new?
Anaplastic thyroid cancer (ATC) is a rare malignancy that is notoriously aggressive. Fatality from ATC remains high, despite advances in multimodal therapy. Here, the authors investigated combination therapy employing tyrosine kinase inhibitors and checkpoint inhibitors as a novel treatment strategy in ATC. In a mouse model of orthotopic ATC, combined treatment with the tyrosine kinase inhibitor lenvatinib and an anti-PD-1/PD-L1 checkpoint inhibitor was found to dramatically reduce tumor volume and improve survival. While lenvatinib monotherapy was associated with increased tumor-infiltrating and circulating myeloid-derived suppressor cells (MDSCs), the depletion of MDSCs via combination therapy significantly augmented lenvatinib's effectiveness.
Supporting Information
Filename | Description |
---|---|
ijc32041-sup-0001-supinfo.docWord document, 676 KB | Appendix S1: Supporting Information |
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.
References
- 1Kebebew E, Greenspan FS, Clark OH, et al. Anaplastic thyroid carcinoma: Treatment outcome and prognostic factors. Cancer 2005; 103: 1330–5.
- 2Prasongsook N, Kumar A, Chintakuntlawar AV, et al. Survival in response to multimodal therapy in anaplastic thyroid cancer. J Clin Endocrinol Metab 2017; 102: 4506–14.
- 3Landa I, Ibrahimpasic T, Boucai L, et al. Genomic and transcriptomic hallmarks of poorly differentiated and anaplastic thyroid cancers. J Clin Invest 2016; 126: 1052–66.
- 4French JD, Bible K, Spitzweg C, et al. Leveraging the immune system to treat advanced thyroid cancers. Lancet Diabetes Endocrinol 2017; 5: 469–81.
- 5Suzuki S, Shibata M, Gonda K, et al. Immunosuppression involving increased myeloid-derived suppressor cell levels, systemic inflammation and hypoalbuminemia are present in patients with anaplastic thyroid cancer. Mol Clin Oncol 2013; 1: 959–64.
- 6Galdiero MR, Varricchi G, Marone G. The immune network in thyroid cancer. Oncoimmunology 2016; 5: e1168556.
- 7Li YL, Zhao H, Ren XB. Relationship of VEGF/VEGFR with immune and cancer cells: staggering or forward? Cancer Biol Med 2016; 13: 206–14.
- 8Voron T, Colussi O, Marcheteau E, et al. VEGF-A modulates expression of inhibitory checkpoints on CD8+ T cells in tumors. J Exp Med 2015; 212: 139–48.
- 9Iyer P, Dadu R, Ferrarotto R, et al. Real world experience with targeted therapy for the treatment of anaplastic thyroid carcinoma. Thyroid 2018; 28: 79–87.
- 10Bible KC, Ryder M. Evolving molecularly targeted therapies for advanced-stage thyroid cancers. Nat Rev Clin Oncol 2016; 13: 403–16.
- 11Cabanillas ME, Habra MA. Lenvatinib: role in thyroid cancer and other solid tumors. Cancer Treat Rev 2016; 42: 47–55.
- 12Schlumberger M, Tahara M, Wirth LJ, et al. Lenvatinib versus placebo in radioiodine-refractory thyroid cancer. N Engl J Med 2015; 372: 621–30.
- 13Tahara M, Kiyota N, Yamazaki T, et al. Lenvatinib for anaplastic thyroid cancer. Front Oncol 2017; 7: 25.
- 14Jing C, Gao Z, Wang R, et al. Lenvatinib enhances the antitumor effects of paclitaxel in anaplastic thyroid cancer. Am J Cancer Res 2017; 7: 903–12.
- 15Huang Y, Goel S, Duda DG, et al. Vascular normalization as an emerging strategy to enhance cancer immunotherapy. Cancer Res 2013; 73: 2943–8.
- 16Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012; 12: 252–64.
- 17Mehnert J. Pembrolizumab for advanced papillary or follicular thyroid cancer: preliminary results from the phase 1b KEYNOTE-028 study. J Clin Oncol 2016; 34: abstr 6091.
- 18Vanden Borre P, McFadden DG, Gunda V, et al. The next generation of orthotopic thyroid cancer models: immunocompetent orthotopic mouse models of BRAF V600E-positive papillary and anaplastic thyroid carcinoma. Thyroid 2014; 24: 705–14.
- 19Brauner E, Gunda V, Vanden Borre P, et al. Combining BRAF inhibitor and anti PD-L1 antibody dramatically improves tumor regression and anti tumor immunity in an immunocompetent murine model of anaplastic thyroid cancer. Oncotarget 2016; 7: 17194–211.
- 20Rodig N, Ryan T, Allen JA, et al. Endothelial expression of PD-L1 and PD-L2 down-regulates CD8+ T cell activation and cytolysis. Eur J Immunol 2003; 33: 3117–26.
- 21Srivastava MK, Zhu L, Harris-White M, et al. Myeloid suppressor cell depletion augments antitumor activity in lung cancer. PLoS One 2012; 7: e40677.
- 22Bray NL, Pimentel H, Melsted P, et al. Near-optimal probabilistic RNA-seq quantification. Nat Biotechnol 2016; 34: 525–7.
- 23Aken BL, Achuthan P, Akanni W, et al. Ensembl 2017. Nucleic Acids Res 2017; 45: D635–D42.
- 24Robinson MD, McCarthy DJ, Smyth GK. edgeR: a bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 2010; 26: 139–40.
- 25Subramanian A, Tamayo P, Mootha VK, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A 2005; 102: 15545–50.
- 26Tomayko MM, Reynolds CP. Determination of subcutaneous tumor size in athymic (nude) mice. Cancer Chemother Pharmacol 1989; 24: 148–54.
- 27Okamoto K, Kodama K, Takase K, et al. Antitumor activities of the targeted multi-tyrosine kinase inhibitor lenvatinib (E7080) against RET gene fusion-driven tumor models. Cancer Lett 2013; 340: 97–103.
- 28Tohyama O, Matsui J, Kodama K, et al. Antitumor activity of lenvatinib (e7080): an angiogenesis inhibitor that targets multiple receptor tyrosine kinases in preclinical human thyroid cancer models. J Thyroid Res 2014; 2014: 638747.
- 29Vivier E, Raulet DH, Moretta A, et al. Innate or adaptive immunity? The example of natural killer cells. Science 2011; 331: 44–9.
- 30Mantovani A, Marchesi F, Malesci A, et al. Tumour-associated macrophages as treatment targets in oncology. Nat Rev Clin Oncol 2017; 14: 399–416.
- 31Ryder M, Ghossein RA, Ricarte-Filho JC, et al. Increased density of tumor-associated macrophages is associated with decreased survival in advanced thyroid cancer. Endocr Relat Cancer 2008; 15: 1069–74.
- 32Bronte V, Brandau S, Chen SH, et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat Commun 2016; 7: 12150.
- 33Martinez FO, Helming L, Gordon S. Alternative activation of macrophages: an immunologic functional perspective. Annu Rev Immunol 2009; 27: 451–83.
- 34Highfill SL, Rodriguez PC, Zhou Q, et al. Bone marrow myeloid-derived suppressor cells (MDSCs) inhibit graft-versus-host disease (GVHD) via an arginase-1-dependent mechanism that is up-regulated by interleukin-13. Blood 2010; 116: 5738–47.
- 35Ward-Hartstonge KA, Kemp RA. Regulatory T-cell heterogeneity and the cancer immune response. Clin Transl Immunol 2017; 6: e154.
- 36Wennerberg E, Pfefferle A, Ekblad L, et al. Human anaplastic thyroid carcinoma cells are sensitive to NK cell-mediated lysis via ULBP2/5/6 and chemoattract NK cells. Clin Cancer Res 2014; 20: 5733–44.
- 37Wang XY, Yi H, Li J. Response to: 'Issues with anti-Gr1 antibody-mediated myeloid-derived suppressor cell depletion' by Xing et al. Ann Rheum Dis 2016; 75: e50.
- 38Lv M, Zhao XS, Hu Y, et al. Monocytic and promyelocytic myeloid-derived suppressor cells may contribute to G-CSF-induced immune tolerance in haplo-identical allogeneic hematopoietic stem cell transplantation. Am J Hematol 2015; 90: E9–E16.
- 39Francisco LM, Salinas VH, Brown KE, et al. PD-L1 regulates the development, maintenance, and function of induced regulatory T cells. J Exp Med 2009; 206: 3015–29.
- 40Umansky V, Blattner C, Gebhardt C, et al. The role of myeloid-derived suppressor cells (MDSC) in cancer progression. Vaccines (Basel) 2016; 4: 1–16.
- 41Angell TE, Lechner MG, Smith AM, et al. Circulating myeloid-derived suppressor cells predict differentiated thyroid cancer diagnosis and extent. Thyroid 2016; 26: 381–9.
- 42Brodaczewska KK, Szczylik C, Kieda C. Immune consequences of anti-angiogenic therapyin renal cell carcinoma. Contemp Oncol (Pozn) 2018; 22: 14–22.
- 43Ostrand-Rosenberg S, Fenselau C. Myeloid-derived suppressor cells: immune-suppressive cells that impair antitumor immunity and are sculpted by their environment. J Immunol 2018; 200: 422–31.
- 44Holmgaard RB, Zamarin D, Lesokhin A, et al. Targeting myeloid-derived suppressor cells with colony stimulating factor-1 receptor blockade can reverse immune resistance to immunotherapy in indoleamine 2,3-dioxygenase-expressing tumors. EBioMedicine 2016; 6: 50–8.
- 45Ko JS, Rayman P, Ireland J, et al. Direct and differential suppression of myeloid-derived suppressor cell subsets by sunitinib is compartmentally constrained. Cancer Res 2010; 70: 3526–36.
- 46Carr LL, Mankoff DA, Goulart BH, et al. Phase II study of daily sunitinib in FDG-PET-positive, iodine-refractory differentiated thyroid cancer and metastatic medullary carcinoma of the thyroid with functional imaging correlation. Clin Cancer Res 2010; 16: 5260–8.
- 47Yu GT, Bu LL, Huang CF, et al. PD-1 blockade attenuates immunosuppressive myeloid cells due to inhibition of CD47/SIRPalpha axis in HPV negative head and neck squamous cell carcinoma. Oncotarget 2015; 6: 42067–80.
- 48Davis RJ, Moore EC, Clavijo PE, et al. Anti-PD-L1 efficacy can be enhanced by inhibition of myeloid-derived suppressor cells with a selective inhibitor of PI3Kdelta/gamma. Cancer Res 2017; 77: 2607–19.