Cryoablation and Immunotherapy: An Enthralling Synergy to Confront the Tumors

Chakradhar Yakkala, Cheryl Lai-Lai Chiang, Lana Kandalaft, Alban Denys, Rafael Duran, Chakradhar Yakkala, Cheryl Lai-Lai Chiang, Lana Kandalaft, Alban Denys, Rafael Duran

Abstract

Treatment of solid tumors by ablation techniques has gained momentum in the recent years due to their technical simplicity and reduced morbidity as juxtaposed to surgery. Cryoablation is one of such techniques, known for its uniqueness to destroy the tumors by freezing to lethal temperatures. Freezing the tumor locally and allowing it to remain in situ unleashes an array of tumor antigens to be exposed to the immune system, paving the way for the generation of anti-tumor immune responses. However, the immune responses triggered in most cases are insufficient to eradicate the tumors with systemic spread. Therefore, combination of cryoablation and immunotherapy is a new treatment strategy currently being evaluated for its efficacy, notably in patients with metastatic disease. This article examines the mechanistic fabric of cryoablation for the generation of an effective immune response against the tumors, and various possibilities of its combination with different immunotherapies that are capable of inducing exceptional therapeutic responses. The combinatorial treatment avenues discussed in this article if explored in sufficient profundity, could reach the pinnacle of future cancer medicine.

Keywords: ablation; cryo; freezing; immunotherapy; treatment; tumors.

Copyright © 2019 Yakkala, Chiang, Kandalaft, Denys and Duran.

Figures

Figure 1
Figure 1
Mechanisms of cell death and immunologic responses induced by cryoablation. (A) Cells in the core of the ablation zone are subjected to lethal temperatures at rapid freezing rates, resulting in the generation of extra and intracellular ice crystals. Cells adjacent to the core zone undergo moderate or low freezing rates. This permits the cells to lose intracellular water by exosmosis in response to the formation of extracellular ice crystals resulting in cellular dehydration and shrinkage. In contrast, cells in the core zone cannot undergo exosmosis due to rapid freezing rates and thus, form intracellular crystals. Both intra and extracellular ice crystals cause mechanical damage to the cells. (B) During the thawing phase, the small intracellular ice crystals, due to their thermodynamic instability, fuse to form larger intracellular crystals (re-crystallization) that enhances the mechanical damage to the cell membranes and intracellular organelles. (C) Post-thawing, mechanically damaged cells die by necrosis and release their contents into the surrounding milieu. Cells that have undergone exosmosis swell and burst due to osmotic shock. Cells in the utmost periphery of the ablation zone exposed to sub-lethal temperatures undergo apoptosis, releasing apoptotic bodies. Antigens released from necrotic cells upon uptake by antigen presenting cells like DCs, induce co-stimulatory signals that would result in the generation of anti-tumoral T-cell responses. In contrast, antigen uptake by DCs in the form of apoptotic bodies imprints immune tolerance or anergy on T-cells due to the non-induction of co-stimulatory signals on DCs. DC, Dendritic cell.
Figure 2
Figure 2
Cryo-immunotherapy: the patient with metastases is treated with local cryoablation of one of them combined with systemic immunotherapies. (A) Administration of low dose toll like receptor (TLR) agonists will cause the activation and maturation of DCs. (B) Cryoablation of a tumor induces tumor necrosis and release of tumor antigens into the surrounding milieu, which are taken up by mature DCs located near the tumor or TDLN. These DCs present tumor antigens to naive T-cells in the TDLN in the presence of agonistic antibodies (for example, CD27) leading to enhanced activation and differentiation into effector T-cells. (C) The effector T-cells thus generated will migrate to the cryoablated tumor site encountering the tumor cells and DCs presenting tumor antigens on their surface MHC molecules. Introduction of checkpoint inhibitors (for example, anti-CTLA-4 and anti-PD-1) will allow the T-cells to execute tumor cell killing without being inhibited by the checkpoint signaling. Eventually, the effector T-cells with blocked checkpoint molecules will also migrate to the distant metastasized tumor sites, leading to the regression of metastases. TLR, toll like receptor; DC, dendritic cell; TDLN, tumor draining lymph node; TCR, T-cell receptor; MHC, major histocompatibility complex.

References

    1. Escudier B, Porta C, Schmidinger M, Rioux-Leclercq N, Bex A, Khoo V, et al. . Renal cell carcinoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. (2016) 27:v58–68. 10.1093/annonc/mdw328
    1. European Association for the Study of the Liver Electronic address eee, European Association for the Study of the Liver. EASL Clinical Practice Guidelines: management of hepatocellular carcinoma. J Hepatol. (2018) 69:182–236. 10.1016/j.jhep.2018.03.024
    1. Chapman WC, Debelak JP, Wright Pinson C, Washington MK, Atkinson JB, Venkatakrishnan A, et al. Hepatic cryoablation, but not radiofrequency ablation, results in lung inflammation. Ann Surg. (2000) 231:752–61. 10.1097/00000658-200005000-00016
    1. Jansen MC, van Hillegersberg R, Schoots IG, Levi M, Beek JF, Crezee H, et al. Cryoablation induces greater inflammatory and coagulative responses than radiofrequency ablation or laser induced thermotherapy in a rat liver model. Surgery. (2010) 147:686–95. 10.1016/j.surg.2009.10.053
    1. Erinjeri JP, Thomas CT, Samoilia A, Fleisher M, Gonen M, Sofocleous CT, et al. . Image-guided thermal ablation of tumors increases the plasma level of interleukin-6 and interleukin-10. J Vasc Interv Radiol. (2013) 24:1105–12. 10.1016/j.jvir.2013.02.015
    1. Shao Q, O'Flanagan S, Lam T, Roy P, Pelaez F, Burbach BJ, et al. . Engineering T cell response to cancer antigens by choice of focal therapeutic conditions. Int J Hyperthermia. (2019) 36:130–8. 10.1080/02656736.2018.1539253
    1. Gage AA, Baust J. Mechanisms of tissue injury in cryosurgery. Cryobiology. (1998) 37:171–86. 10.1006/cryo.1998.2115
    1. Sidana A, Chowdhury WH, Fuchs EJ, Rodriguez R. Cryoimmunotherapy in urologic oncology. Urology. (2010) 75:1009–14. 10.1016/j.urology.2009.06.020
    1. Korpan NN. A history of cryosurgery: its development and future. J Am Coll Surg. (2007) 204:314–24. 10.1016/j.jamcollsurg.2006.11.006
    1. Cooper SM, Dawber RP. The history of cryosurgery. J R Soc Med. (2001) 94:196–201. 10.1177/014107680109400416
    1. Allington HV. Liquid nitrogen in the treatment of skin diseases. Calif Med. (1950) 72:153–5.
    1. Rowbotham GF, Haigh AL, Leslie WG. Cooling cannula for use in the treatment of cerebral neoplasms. Lancet. (1959) 1:12–5. 10.1016/S0140-6736(59)90976-6
    1. Cooper IS, Lee AS. Cryostatic congelation: a system for producing a limited, controlled region of cooling or freezing of biologic tissues. J Nerv Ment Dis. (1961) 133:259–63. 10.1097/00005053-196109000-00013
    1. Baust JG, Gage AA, Bjerklund Johansen TE, Baust JM. Mechanisms of cryoablation: clinical consequences on malignant tumors. Cryobiology. (2014) 68:1–11. 10.1016/j.cryobiol.2013.11.001
    1. Erinjeri JP, Clark TW. Cryoablation: mechanism of action and devices. J Vasc Interv Radiol. (2010) 21:S187–91. 10.1016/j.jvir.2009.12.403
    1. Gage AA, Baust JM, Baust JG. Experimental cryosurgery investigations in vivo. Cryobiology. (2009) 59:229–43. 10.1016/j.cryobiol.2009.10.001
    1. Mazur P. Freezing of living cells: mechanisms and implications. Am J Physiol. (1984) 247:C125–42. 10.1152/ajpcell.1984.247.3.C125
    1. Hoffmann NE, Bischof JC. The cryobiology of cryosurgical injury. Urology. (2002) 60:40–9. 10.1016/S0090-4295(02)01683-7
    1. Bryant G. DSC measurement of cell suspensions during successive freezing runs: implications for the mechanisms of intracellular ice formation. Cryobiology. (1995) 32:114–28. 10.1006/cryo.1995.1011
    1. Mazur P. Kinetics of water loss from cells at subzero temperatures and the likelihood of intracellular freezing. J Gen Physiol. (1963) 47:347–69. 10.1085/jgp.47.2.347
    1. Gage AA, Meenaghan MA, Natiella JR, Greene GW, Jr. Sensitivity of pigmented mucosa and skin to freezing injury. Cryobiology. (1979) 16:348–61. 10.1016/0011-2240(79)90048-8
    1. Baust JG, Gage AA. Progress toward optimization of cryosurgery. Technol Cancer Res Treat. (2004) 3:95–101. 10.1177/153303460400300202
    1. Zhou T, Li Q, Chi X, Li L, Rao J, Hu K. 0 degrees C is better?- Thawing temperature optimization study for cancer cryoablation in a mouse model with green fluorescent protein-labeled Lewis lung cancer. Cryobiology. (2017) 75:80–7. 10.1016/j.cryobiol.2017.01.008
    1. Sabel MS. Cryo-immunology: a review of the literature and proposed mechanisms for stimulatory versus suppressive immune responses. Cryobiology. (2009) 58:1–11. 10.1016/j.cryobiol.2008.10.126
    1. Theodorescu D. Cancer cryotherapy: evolution and biology. Rev Urol. (2004) 6(Suppl. 4):S9–19. 10.1186/1475-2867-4-S1-S19
    1. Takahashi Y, Izumi Y, Matsutani N, Dejima H, Nakayama T, Okamura R, et al. . Optimized magnitude of cryosurgery facilitating anti-tumor immunoreaction in a mouse model of Lewis lung cancer. Cancer Immunol Immunother. (2016) 65:973–82. 10.1007/s00262-016-1858-x
    1. Chu KF, Dupuy DE. Thermal ablation of tumours: biological mechanisms and advances in therapy. Nat Rev Cancer. (2014) 14:199–208. 10.1038/nrc3672
    1. Maccini M, Sehrt D, Pompeo A, Chicoli FA, Molina WR, Kim FJ. Biophysiologic considerations in cryoablation: a practical mechanistic molecular review. Int Braz J Urol. (2011) 37:693–6. 10.1590/S1677-55382011000600002
    1. Alblin RJ, Soanes WA, Gonder MJ. Prospects for cryo-immunotherapy in cases of metastasizing carcinoma of the prostate. Cryobiology. (1971) 8:271–9. 10.1016/0011-2240(71)90050-2
    1. Gursel E, Roberts M, Veenema RJ. Regression of prostatic cancer following sequential cryotherapy to the prostate. J Urol. (1972) 108:928–32. 10.1016/S0022-5347(17)60909-1
    1. Soanes WA, Ablin RJ, Gonder MJ. Remission of metastatic lesions following cryosurgery in prostatic cancer: immunologic considerations. J Urol. (1970) 104:154–9. 10.1016/S0022-5347(17)61690-2
    1. Uhlschmid G, Kolb E, Largiader F. Cryosurgery of pulmonary metastases. Cryobiology. (1979) 16:171–8. 10.1016/0011-2240(79)90028-2
    1. Misao A, Sakata K, Saji S, Kunieda T. Late appearance of resistance to tumor rechallenge following cryosurgery. A study in an experimental mammary tumor of the rat. Cryobiology. (1981) 18:386–9. 10.1016/0011-2240(81)90111-5
    1. Miya K, Saji S, Morita T, Niwa H, Takao H, Kida H, et al. . Immunological response of regional lymph nodes after tumor cryosurgery: experimental study in rats. Cryobiology. (1986) 23:290–5. 10.1016/0011-2240(86)90034-9
    1. Neel HB, 3rd, Ketcham AS, Hammond WG. Experimental evaluation of in situ oncocide for primary tumor therapy: comparison of tumor-specific immunity after complete excision, cryonecrosis and ligation. Laryngoscope. (1973) 83:376–87. 10.1288/00005537-197303000-00009
    1. Sabel MS, Nehs MA, Su G, Lowler KP, Ferrara JL, Chang AE. Immunologic response to cryoablation of breast cancer. Breast Cancer Res Treat. (2005) 90:97–104. 10.1007/s10549-004-3289-1
    1. Sabel MS, Arora A, Su G, Chang AE. Adoptive immunotherapy of breast cancer with lymph node cells primed by cryoablation of the primary tumor. Cryobiology. (2006) 53:360–6. 10.1016/j.cryobiol.2006.07.004
    1. den Brok MH, Sutmuller RP, Nierkens S, Bennink EJ, Toonen LW, Figdor CG, et al. . Synergy between in situ cryoablation and TLR9 stimulation results in a highly effective in vivo dendritic cell vaccine. Cancer Res. (2006) 66:7285–92. 10.1158/0008-5472.CAN-06-0206
    1. Osada S, Imai H, Tomita H, Tokuyama Y, Okumura N, Matsuhashi N, et al. . Serum cytokine levels in response to hepatic cryoablation. J Surg Oncol. (2007) 95:491–8. 10.1002/jso.20712
    1. Beatty GL, Gladney WL. Immune escape mechanisms as a guide for cancer immunotherapy. Clin Cancer Res. (2015) 21:687–92. 10.1158/1078-0432.CCR-14-1860
    1. Otto G. Tumour decides immune cell ins and outs. Nat Rev Immunol. (2018) 18:481. 10.1038/s41577-018-0038-y
    1. Seton-Rogers S. Oncogenes: driving immune evasion. Nat Rev Cancer. (2018) 18:67. 10.1038/nrc.2018.5
    1. Kaczanowska S, Joseph AM, Davila E. TLR agonists: our best frenemy in cancer immunotherapy. J Leukoc Biol. (2013) 93:847–63. 10.1189/jlb.1012501
    1. Kawasaki T, Kawai T. Toll-like receptor signaling pathways. Front Immunol. (2014) 5:461. 10.3389/fimmu.2014.00461
    1. Redondo P, del Olmo J, Lopez-Diaz de Cerio A, Inoges S, Marquina M, Melero I, et al. . Imiquimod enhances the systemic immunity attained by local cryosurgery destruction of melanoma lesions. J Invest Dermatol. (2007) 127:1673–80. 10.1038/sj.jid.5700777
    1. Gaitanis G, Nomikos K, Vava E, Alexopoulos EC, Bassukas ID. Immunocryosurgery for basal cell carcinoma: results of a pilot, prospective, open-label study of cryosurgery during continued imiquimod application. J Eur Acad Dermatol Venereol. (2009) 23:1427–31. 10.1111/j.1468-3083.2009.03224.x
    1. Merad M, Sathe P, Helft J, Miller J, Mortha A. The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu Rev Immunol. (2013) 31:563–604. 10.1146/annurev-immunol-020711-074950
    1. Gardner A, Ruffell B. Dendritic cells and cancer immunity. Trends Immunol. (2016) 37:855–65. 10.1016/j.it.2016.09.006
    1. Bevan MJ. Cross-priming for a secondary cytotoxic response to minor H antigens with H-2 congenic cells which do not cross-react in the cytotoxic assay. J Exp Med. (1976) 143:1283–8. 10.1084/jem.143.5.1283
    1. Garg AD, Coulie PG, Van den Eynde BJ, Agostinis P. Integrating next-generation dendritic cell vaccines into the current cancer immunotherapy landscape. Trends Immunol. (2017) 38:577–93. 10.1016/j.it.2017.05.006
    1. Constantino J, Gomes C, Falcao A, Cruz MT, Neves BM. Antitumor dendritic cell-based vaccines: lessons from 20 years of clinical trials and future perspectives. Transl Res. (2016) 168:74–95. 10.1016/j.trsl.2015.07.008
    1. Anguille S, Smits EL, Lion E, van Tendeloo VF, Berneman ZN. Clinical use of dendritic cells for cancer therapy. Lancet Oncol. (2014) 15:e257–67. 10.1016/S1470-2045(13)70585-0
    1. Machlenkin A, Goldberger O, Tirosh B, Paz A, Volovitz I, Bar-Haim E, et al. . Combined dendritic cell cryotherapy of tumor induces systemic antimetastatic immunity. Clin Cancer Res. (2005) 11:4955–61. 10.1158/1078-0432.CCR-04-2422
    1. Udagawa M, Kudo-Saito C, Hasegawa G, Yano K, Yamamoto A, Yaguchi M, et al. . Enhancement of immunologic tumor regression by intratumoral administration of dendritic cells in combination with cryoablative tumor pretreatment and Bacillus Calmette-Guerin cell wall skeleton stimulation. Clin Cancer Res. (2006) 12:7465–75. 10.1158/1078-0432.CCR-06-1840
    1. Lowry LE, Zehring WA. Potentiation of natural killer cells for cancer immunotherapy: a review of literature. Front Immunol. (2017) 8:1061. 10.3389/fimmu.2017.01061
    1. Rezvani K, Rouce R, Liu E, Shpall E. Engineering natural killer cells for cancer immunotherapy. Mol Ther. (2017) 25:1769–81. 10.1016/j.ymthe.2017.06.012
    1. Imai K, Matsuyama S, Miyake S, Suga K, Nakachi K. Natural cytotoxic activity of peripheral-blood lymphocytes and cancer incidence: an 11-year follow-up study of a general population. Lancet. (2000) 356:1795–9. 10.1016/S0140-6736(00)03231-1
    1. Rezvani K, Rouce RH. The application of natural killer cell immunotherapy for the treatment of cancer. Front Immunol. (2015) 6:578. 10.3389/fimmu.2015.00578
    1. Iliopoulou EG, Kountourakis P, Karamouzis MV, Doufexis D, Ardavanis A, Baxevanis CN, et al. . A phase I trial of adoptive transfer of allogeneic natural killer cells in patients with advanced non-small cell lung cancer. Cancer Immunol Immunother. (2010) 59:1781–9. 10.1007/s00262-010-0904-3
    1. Lin M, Liang S, Wang X, Liang Y, Zhang M, Chen J, et al. . Cryoablation combined with allogenic natural killer cell immunotherapy improves the curative effect in patients with advanced hepatocellular cancer. Oncotarget. (2017) 8:81967–77. 10.18632/oncotarget.17804
    1. Lin M, Liang SZ, Wang XH, Liang YQ, Zhang MJ, Niu LZ, et al. . Clinical efficacy of percutaneous cryoablation combined with allogenic NK cell immunotherapy for advanced non-small cell lung cancer. Immunol Res. (2017) 65:880–7. 10.1007/s12026-017-8927-x
    1. Lin M, Xu K, Liang S, Wang X, Liang Y, Zhang M, et al. . Prospective study of percutaneous cryoablation combined with allogenic NK cell immunotherapy for advanced renal cell cancer. Immunol Lett. (2017) 184:98–104. 10.1016/j.imlet.2017.03.004
    1. Gao X, Mi Y, Guo N, Xu H, Xu L, Gou X, et al. . Cytokine-induced killer cells as pharmacological tools for cancer immunotherapy. Front Immunol. (2017) 8:774. 10.3389/fimmu.2017.00774
    1. Liu L, Zhang W, Qi X, Li H, Yu J, Wei S, et al. . Randomized study of autologous cytokine-induced killer cell immunotherapy in metastatic renal carcinoma. Clin Cancer Res. (2012) 18:1751–9. 10.1158/1078-0432.CCR-11-2442
    1. Pan K, Guan XX, Li YQ, Zhao JJ, Li JJ, Qiu HJ, et al. . Clinical activity of adjuvant cytokine-induced killer cell immunotherapy in patients with post-mastectomy triple-negative breast cancer. Clin Cancer Res. (2014) 20:3003–11. 10.1158/1078-0432.CCR-14-0082
    1. Lee JH, Lee JH, Lim YS, Yeon JE, Song TJ, Yu SJ, et al. . Adjuvant immunotherapy with autologous cytokine-induced killer cells for hepatocellular carcinoma. Gastroenterology. (2015) 148:1383–91 e1386. 10.1053/j.gastro.2015.02.055
    1. Niu LZ, Li JL, Zeng JY, Mu F, Liao MT, Yao F, et al. . Combination treatment with comprehensive cryoablation and immunotherapy in metastatic hepatocellular cancer. World J Gastroenterol. (2013) 19:3473–80. 10.3748/wjg.v19.i22.3473
    1. Niu L, Chen J, He L, Liao M, Yuan Y, Zeng J, et al. . Combination treatment with comprehensive cryoablation and immunotherapy in metastatic pancreatic cancer. Pancreas. (2013) 42:1143–9. 10.1097/MPA.0b013e3182965dde
    1. Yuanying Y, Lizhi N, Feng M, Xiaohua W, Jianying Z, Fei Y, et al. . Therapeutic outcomes of combining cryotherapy, chemotherapy and DC-CIK immunotherapy in the treatment of metastatic non-small cell lung cancer. Cryobiology. (2013) 67:235–40. 10.1016/j.cryobiol.2013.08.001
    1. Silva-Santos B, Serre K, Norell H. gammadelta T cells in cancer. Nat Rev Immunol. (2015) 15:683–91. 10.1038/nri3904
    1. Lo Presti E, Pizzolato G, Gulotta E, Cocorullo G, Gulotta G, Dieli F, et al. . Current advances in gammadelta T cell-based tumor immunotherapy. Front Immunol. (2017) 8:1401. 10.3389/fimmu.2017.01401
    1. Pauza CD, Liou ML, Lahusen T, Xiao L, Lapidus RG, Cairo C, et al. . Gamma delta T cell therapy for cancer: it is good to be local. Front Immunol. (2018) 9:1305. 10.3389/fimmu.2018.01305
    1. Gober HJ, Kistowska M, Angman L, Jeno P, Mori L, De Libero G. Human T cell receptor gammadelta cells recognize endogenous mevalonate metabolites in tumor cells. J Exp Med. (2003) 197:163–8. 10.1084/jem.20021500
    1. Chargui J, Combaret V, Scaglione V, Iacono I, Peri V, Valteau-Couanet D, et al. . Bromohydrin pyrophosphate-stimulated Vgamma9delta2 T cells expanded ex vivo from patients with poor-prognosis neuroblastoma lyse autologous primary tumor cells. J Immunother. (2010) 33:591–8. 10.1097/CJI.0b013e3181dda207
    1. Zhou J, Kang N, Cui L, Ba D, He W. Anti-gammadelta TCR antibody-expanded gammadelta T cells: a better choice for the adoptive immunotherapy of lymphoid malignancies. Cell Mol Immunol. (2012) 9:34–44. 10.1038/cmi.2011.16
    1. Brandes M, Willimann K, Moser B. Professional antigen-presentation function by human gammadelta T Cells. Science. (2005) 309:264–8. 10.1126/science.1110267
    1. Fisher JP, Heuijerjans J, Yan M, Gustafsson K, Anderson J. gammadelta T cells for cancer immunotherapy: a systematic review of clinical trials. Oncoimmunology. (2014) 3:e27572. 10.4161/onci.27572
    1. Bennouna J, Bompas E, Neidhardt EM, Rolland F, Philip I, Galea C, et al. . Phase-I study of Innacell gammadelta, an autologous cell-therapy product highly enriched in gamma9delta2 T lymphocytes, in combination with IL-2, in patients with metastatic renal cell carcinoma. Cancer Immunol Immunother. (2008) 57:1599–609. 10.1007/s00262-008-0491-8
    1. Sakamoto M, Nakajima J, Murakawa T, Fukami T, Yoshida Y, Murayama T, et al. Adoptive immunotherapy for advanced non-small cell lung cancer using zoledronate-expanded gammadelta T cells: a phase I clinical study. J Immunother. (2011) 34:202–11. 10.1097/CJI.0b013e318207ecfb
    1. Nicol AJ, Tokuyama H, Mattarollo SR, Hagi T, Suzuki K, Yokokawa K, et al. . Clinical evaluation of autologous gamma delta T cell-based immunotherapy for metastatic solid tumours. Br J Cancer. (2011) 105:778–86. 10.1038/bjc.2011.293
    1. Fleming C, Morrissey S, Cai Y, Yan J. Gammadelta T cells: unexpected regulators of cancer development and progression. Trends Cancer. (2017) 3:561–70. 10.1016/j.trecan.2017.06.003
    1. Parente-Pereira AC, Shmeeda H, Whilding LM, Zambirinis CP, Foster J, van der Stegen SJ, et al. . Adoptive immunotherapy of epithelial ovarian cancer with Vgamma9Vdelta2 T cells, potentiated by liposomal alendronic acid. J Immunol. (2014) 193:5557–66. 10.4049/jimmunol.1402200
    1. Santolaria T, Robard M, Leger A, Catros V, Bonneville M, Scotet E. Repeated systemic administrations of both aminobisphosphonates and human Vgamma9Vdelta2 T cells efficiently control tumor development in vivo. J Immunol. (2013) 191:1993–2000. 10.4049/jimmunol.1300255
    1. de Charette M, Marabelle A, Houot R. Turning tumour cells into antigen presenting cells: the next step to improve cancer immunotherapy? Eur J Cancer. (2016) 68:134–47. 10.1016/j.ejca.2016.09.010
    1. Garrido F, Aptsiauri N, Doorduijn EM, Garcia Lora AM, van Hall T. The urgent need to recover MHC class I in cancers for effective immunotherapy. Curr Opin Immunol. (2016) 39:44–51. 10.1016/j.coi.2015.12.007
    1. Gentles AJ, Newman AM, Liu CL, Bratman SV, Feng W, Kim D, et al. . The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat Med. (2015) 21:938–45. 10.1038/nm.3909
    1. Greenwald RJ, Freeman GJ, Sharpe AH. The B7 family revisited. Annu Rev Immunol. (2005) 23:515–48. 10.1146/annurev.immunol.23.021704.115611
    1. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science. (2018) 359:1350–5. 10.1126/science.aar4060
    1. Zou W, Chen L. Inhibitory B7-family molecules in the tumour microenvironment. Nat Rev Immunol. (2008) 8:467–77. 10.1038/nri2326
    1. Gubin MM, Zhang X, Schuster H, Caron E, Ward JP, Noguchi T, et al. . Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature. (2014) 515:577–81. 10.1038/nature13988
    1. Ribas A. Releasing the brakes on cancer immunotherapy. N Engl J Med. (2015) 373:1490–2. 10.1056/NEJMp1510079
    1. Waitz R, Solomon SB, Petre EN, Trumble AE, Fasso M, Norton L, et al. . Potent induction of tumor immunity by combining tumor cryoablation with anti-CTLA-4 therapy. Cancer Res. (2012) 72:430–9. 10.1158/0008-5472.CAN-11-1782
    1. den Brok MH, Sutmuller RP, Nierkens S, Bennink EJ, Frielink C, Toonen LW, et al. . Efficient loading of dendritic cells following cryo and radiofrequency ablation in combination with immune modulation induces anti-tumour immunity. Br J Cancer. (2006) 95:896–905. 10.1038/sj.bjc.6603341
    1. Kim DW, Haymaker C, McQuail N, Sirmans E, Spencer C, Glitza I, et al. Pilot study of intratumoral (IT) cryoablation (cryo) in combination with systemic checkpoint blockade in patients with metastatic melanoma (MM). J ImmunoTher Cancer. (2015) 3:P137 10.1186/2051-1426-3-S2-P137
    1. McArthur HL, Diab A, Page DB, Yuan J, Solomon SB, Sacchini V, et al. . A pilot study of preoperative single-dose ipilimumab and/or cryoablation in women with early-stage breast cancer with comprehensive immune profiling. Clin Cancer Res. (2016) 22:5729–37. 10.1158/1078-0432.CCR-16-0190
    1. Kato T, Iwasaki T, Uemura M, Nagahara A, Higashihara H, Osuga K, et al. . Characterization of the cryoablation-induced immune response in kidney cancer patients. Oncoimmunology. (2017) 6:e1326441. 10.1080/2162402X.2017.1326441
    1. Glazer DI, Tatli S, Shyn PB, Vangel MG, Tuncali K, Silverman SG. Percutaneous image-guided cryoablation of hepatic tumors: single-center experience with intermediate to long-term outcomes. AJR Am J Roentgenol. (2017) 209:1381–9. 10.2214/AJR.16.17582
    1. Packard AT, Broski SM, Callstrom MR, Atwell TD, Schmit GD, Schmitz JJ, et al. . Utility of PET/CT after cryoablation for early identification of local tumor progression in osseous metastatic disease. AJR Am J Roentgenol. (2017) 208:1342–51. 10.2214/AJR.16.17222
    1. Permpongkosol S, Nicol TL, Khurana H, Link RE, Zhai QJ, Kavoussi LR, et al. . Thermal maps around two adjacent cryoprobes creating overlapping ablations in porcine liver, lung, and kidney. J Vasc Interv Radiol. (2007) 18:283–7. 10.1016/j.jvir.2006.12.008
    1. Permpongkosol S, Nicol TL, Link RE, Varkarakis I, Khurana H, Zhai QJ, et al. . Differences in ablation size in porcine kidney, liver, and lung after cryoablation using the same ablation protocol. AJR Am J Roentgenol. (2007) 188:1028–32. 10.2214/AJR.06.0810
    1. Marin-Acevedo JA, Dholaria B, Soyano AE, Knutson KL, Chumsri S, Lou Y. Next generation of immune checkpoint therapy in cancer: new developments and challenges. J Hematol Oncol. (2018) 11:39. 10.1186/s13045-018-0582-8
    1. Mayes PA, Hance KW, Hoos A. The promise and challenges of immune agonist antibody development in cancer. Nat Rev Drug Discov. (2018) 17:509–27. 10.1038/nrd.2018.75
    1. de Baere T, Tselikas L, Deschamps F, Soria JC, Marabelle A. Immuno-oncology in cancer care is a fantastic opportunity for interventional oncology: IO4IO (Interventional Oncology for Immuno-Oncology) Initiative. Cardiovasc Intervent Radiol. (2018) 41:825–7. 10.1007/s00270-018-1935-y
    1. Marabelle A, Tselikas L, de Baere T, Houot R. Intratumoral immunotherapy: using the tumor as the remedy. Ann Oncol. (2017) 28:xii33–43. 10.1093/annonc/mdx683
    1. Rosenberg MA, Williams J. Image guided cryoablation of cancer with intra-tumoral injection of anti-CTLA-4 and PD-1 immune check-point inhibitors. J ImmunoTher Cancer. (2015) 3:P142 10.1186/2051-1426-3-S2-P142
    1. Haanen J. Converting cold into hot tumors by combining immunotherapies. Cell. (2017) 170:1055–6. 10.1016/j.cell.2017.08.031
    1. Mizukami Y, Kono K, Maruyama T, Watanabe M, Kawaguchi Y, Kamimura K, et al. . Downregulation of HLA Class I molecules in the tumour is associated with a poor prognosis in patients with oesophageal squamous cell carcinoma. Br J Cancer. (2008) 99:1462–7. 10.1038/sj.bjc.6604715
    1. Newick K, O'Brien S, Moon E, Albelda SM. CAR T cell therapy for solid tumors. Annu Rev Med. (2017) 68:139–52. 10.1146/annurev-med-062315-120245
    1. Angelova M, Mlecnik B, Vasaturo A, Bindea G, Fredriksen T, Lafontaine L, et al. . Evolution of metastases in space and time under immune selection. Cell. (2018) 175:751–65 e716. 10.1016/j.cell.2018.09.018

Source: PubMed

3
Abonner