Journal Home > Volume 2 , Issue 5

Local therapy modalities such as radiation therapy, photodynamic therapy, photothermal therapy, and cryoablation have been used to treat localized tumors for decades. The discovery of the abscopal effect causes a paradigm shift where local therapy also causes systemic effects and leads to the remission of nonirradiated tumors. The abscopal effect of radiation therapy, alone or in combination with other treatments, has been extensively studied over the last six decades. However, the results are unsatisfactory in producing robust, reproducible, and long‐lasting systemic effects. Although immunotherapy and radiation therapy are promising in producing the abscopal effect, the abscopal effect's mechanism is still unclear, owing to various factors such as irradiation type and dose and cancer type. This article reviews the research progress, clinical and preclinical evidence of the abscopal effect by various local therapies alone and in combination with chemotherapy and immunotherapy, case reports, and the current challenges in producing the abscopal effect by various local therapies, focusing on radiotherapy, photodynamic therapy, cryoablation, and the prospects for obtaining a robust, reproducible, and long‐lasting abscopal effect.


menu
Abstract
Full text
Outline
About this article

Radiotherapy, photodynamic therapy, and cryoablation‐induced abscopal effect: Challenges and future prospects

Show Author's information Sadik Ali MohammadArshadul HakSunil V. PoguAravind K. Rengan ( )
Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana India

Abstract

Local therapy modalities such as radiation therapy, photodynamic therapy, photothermal therapy, and cryoablation have been used to treat localized tumors for decades. The discovery of the abscopal effect causes a paradigm shift where local therapy also causes systemic effects and leads to the remission of nonirradiated tumors. The abscopal effect of radiation therapy, alone or in combination with other treatments, has been extensively studied over the last six decades. However, the results are unsatisfactory in producing robust, reproducible, and long‐lasting systemic effects. Although immunotherapy and radiation therapy are promising in producing the abscopal effect, the abscopal effect's mechanism is still unclear, owing to various factors such as irradiation type and dose and cancer type. This article reviews the research progress, clinical and preclinical evidence of the abscopal effect by various local therapies alone and in combination with chemotherapy and immunotherapy, case reports, and the current challenges in producing the abscopal effect by various local therapies, focusing on radiotherapy, photodynamic therapy, cryoablation, and the prospects for obtaining a robust, reproducible, and long‐lasting abscopal effect.

Keywords: immunotherapy, photodynamic therapy, cancer, radiotherapy, abscopal effect, local therapy, cryoablation

References(176)

1

Mole RH. Whole body irradiation; radiobiology or medicine? Br J Radiol. 1953;26(305):234–41. https://doi.org/10.1259/0007-1285-26-305-234

2

Ribeiro Gomes J, Schmerling RA, Haddad CK, Racy DJ, Ferrigno R, Gil E, et al. Analysis of the abscopal effect with anti‐PD1 therapy in patients with metastatic solid tumors. J Immunother. 2016;39(9):367–72. https://doi.org/10.1097/CJI.0000000000000141

3

Postow MA, Callahan MK, Barker CA, Yamada Y, Yuan J, Kitano S, et al. Immunologic correlates of the abscopal effect in a patient with melanoma. N Engl J Med. 2012;366(10):925–31. https://doi.org/10.1056/nejmoa1112824

4

Deng L, Liang H, Burnette B, Beckett M, Darga T, Weichselbaum RR, et al. Irradiation and anti‐PD‐L1 treatment synergistically promote antitumor immunity in mice. J Clin Invest. 2014;124:687–95. https://doi.org/10.1172/JCI67313

5

Dovedi SJ, Adlard AL, Lipowska‐Bhalla G, McKenna C, Jones S, Cheadle EJ, et al. Acquired resistance to fractionated radiotherapy can be overcome by concurrent PD‐L1 blockade. Cancer Res. 2014;74(19):5458–68. https://doi.org/10.1158/0008-5472.CAN-14-1258

6

Demaria S, Ng B, Devitt ML, Babb JS, Kawashima N, Liebes L, et al. Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated. Int J Radiat Oncol Biol Phys. 2004;58:862–70. https://doi.org/10.1016/j.ijrobp.2003.09.012

7

Sia J, Szmyd R, Hau E, Gee HE. Molecular mechanisms of radiation‐induced cancer cell death: a primer. Front Cell Dev Biol. 2020;8:41. https://doi.org/10.3389/fcell.2020.00041

8

Nakajima K, Shibamoto Y, Kobayashi M, Takaoka T, Murai T, Manabe Y, et al. The abscopal effect in patients with multiple metastases treated with combination of dendritic cell‐based immunotherapy and focal radiation therapy. Int J Radiat Oncol Biol Phys. 2016;96(2):E570–71. https://doi.org/10.1016/j.ijrobp.2016.06.2056

9

Demaria S, Kawashima N, Yang AM, Devitt ML, Babb JS, Allison JP, et al. Immune‐mediated inhibition of metastases after treatment with local radiation and CTLA‐4 blockade in a mouse model of breast cancer. Clin Cancer Res. 2005;11(2):728–34. https://doi.org/10.1158/1078-0432.728.11.2

10

Shi F, Wang X, Teng F, Kong L, Yu J. Abscopal effect of metastatic pancreatic cancer after local radiotherapy and granulocyte–macrophage colony‐stimulating factor therapy. Cancer Biol Ther. 2017;18(3):137–41. https://doi.org/10.1080/15384047.2016.1276133

11

Nakajima N, Kano T, Oda K, Uchida T, Otaki T, Nagao K, et al. Possible abscopal effect after discontinuation of nivolumab in metastatic renal cell carcinoma. IJU Case Rep. 2020;3:215–8. https://doi.org/10.1002/iju5.12195

12

Havunen R, Santos JM, Sorsa S, Rantapero T, Lumen D, Siurala M, et al. Abscopal effect in non‐injected tumors achieved with cytokine‐armed oncolytic adenovirus. Mol Ther Oncol. 2018;11:109–21. https://doi.org/10.1016/j.omto.2018.10.005

13

Kwon ED, Drake CG, Scher HI, Fizazi K, Bossi A, Van den Eertwegh AJM, et al. Ipilimumab versus placebo after radiotherapy in patients with metastatic castration‐resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184‐043): a multicentre, randomised, double‐blind, phase 3 trial. Lancet Oncol. 2014;15(7):700–12. https://doi.org/10.1016/S1470-2045(14)70189-5

14

Thong PSP, Ong KW, Goh NSG, Kho KW, Manivasager V, Bhuvaneswari R, et al. Photodynamic‐therapy‐activated immune response against distant untreated tumours in recurrent angiosarcoma. Lancet Oncol. 2007;8(10):950–2. https://doi.org/10.1016/S1470-2045(07)70318-2

15

Song W, Kuang J, Li C‐X, Zhang M, Zheng D, Zeng X, et al. Enhanced immunotherapy based on photodynamic therapy for both primary and lung metastasis tumor eradication. ACS Nano. 2018;12(2):1978–89. https://doi.org/10.1021/acsnano.7b09112

16

Cano‐Mejia J, Shukla A, Ledezma DK, Palmer E, Villagra A, Fernandes R. CpG‐coated Prussian blue nanoparticles‐based photothermal therapy combined with anti‐CTLA‐4 immune checkpoint blockade triggers a robust abscopal effect against neuroblastoma. Transl Oncol. 2020;13(10):100823. https://doi.org/10.1016/j.tranon.2020.100823

17

Soule E, Bandyk M, Matteo J. Percutaneous ablative cryoimmunotherapy for micrometastaic abscopal effect: no complications. Cryobiology. 2018;82:22–6. https://doi.org/10.1016/j.cryobiol.2018.04.013

18

Eranki A, Srinivasan P, Ries M, Kim A, Lazarski CA, Rossi CT, et al. High‐intensity focused ultrasound (hIFU) triggers immune sensitization of refractory murine neuroblastoma to checkpoint inhibitor therapy. Clin Cancer Res. 2020;26:1152–61. https://doi.org/10.1158/1078-0432.CCR-19-1604

19

Chi MS, Mehta MP, Yang KL, Lai HC, Lin YC, Ko HL, et al. Putative abscopal effect in three patients treated by combined radiotherapy and modulated electrohyperthermia. Front Oncol. 2020;10:254. https://doi.org/10.3389/fonc.2020.00254

20

Galluzzi L, Buqué A, Kepp O, Zitvogel L, Kroemer G. Immunogenic cell death in cancer and infectious disease. Nat Rev Immunol. 2017;17(2):97–111. https://doi.org/10.1038/nri.2016.107

21

Korbelik M, Zhang W, Merchant S. Involvement of damage‐associated molecular patterns in tumor response to photodynamic therapy: surface expression of calreticulin and high‐mobility group box‐1 release. Cancer Immunol Immunother. 2011;60(10):1431–7. https://doi.org/10.1007/s00262-011-1047-x

22

Stone HB, Peters LJ, Milas L. Effect of host immune capability on radiocurability and subsequent transplantability of a murine fibrosarcoma. J Natl Cancer Inst. 1979;63(5):1229–35. https://doi.org/10.1002/ijc.2910140409

23

Rodriguez‐Ruiz ME, Rodriguez I, Leaman O, López‐Campos F, Montero A, Conde AJ, et al. Immune mechanisms mediating abscopal effects in radioimmunotherapy. Pharmacol Ther. 2019;196:195–203. https://doi.org/10.1016/j.pharmthera.2018.12.002

24

Korbelik M. Role of cell stress signaling networks in cancer cell death and antitumor immune response following proteotoxic injury inflicted by photodynamic therapy. Lasers Surg Med. 2018;50(5):491–8. https://doi.org/10.1002/lsm.22810

25

Obeid M. ERP57 membrane translocation dictates the immunogenicity of tumor cell death by controlling the membrane translocation of calreticulin. J Immunol. 2008;181(4):2533–43. https://doi.org/10.4049/jimmunol.181.4.2533

26

Sims GP, Rowe DC, Rietdijk ST, Herbst R, Coyle AJ. HMGB1 and RAGE in inflammation and cancer. Annu Rev Immunol. 2010;28:367–88. https://doi.org/10.1146/annurev.immunol.021908.132603

27

Turubanova VD, Balalaeva Iv, Mishchenko TA, Catanzaro E, Alzeibak R, Peskova NN, et al. Immunogenic cell death induced by a new photodynamic therapy based on photosens and photodithazine. J Immunother Cancer. 2019;7(1):350. https://doi.org/10.1186/s40425-019-0826-3

28

Marill J, Mohamed Anesary N, Paris S. DNA damage enhancement by radiotherapy‐activated hafnium oxide nanoparticles improves cGAS‐STING pathway activation in human colorectal cancer cells. Radiother Oncol. 2019;141:262–6. https://doi.org/10.1016/j.radonc.2019.07.029

29

Grass GD, Krishna N, Kim S. The immune mechanisms of abscopal effect in radiation therapy. Curr Probl Cancer. 2016;40(1):10–24. https://doi.org/10.1016/j.currproblcancer.2015.10.003

30

Formenti SC, Demaria S. Combining radiotherapy and cancer immunotherapy: a paradigm shift. J Natl Cancer Inst. 2013;105(4):256–65. https://doi.org/10.1093/jnci/djs629

31

Lugade AA, Sorensen EW, Gerber SA, Moran JP, Frelinger JG, Lord EM. Radiation‐induced IFN‐γ production within the tumor microenvironment influences antitumor immunity. J Immunol. 2008;180(5):3132–9. https://doi.org/10.4049/jimmunol.180.5.3132

32

Lim JYH, Gerber SA, Murphy SP, Lord EM. Type Ⅰ interferons induced by radiation therapy mediate recruitment and effector function of CD8+ T cells. Cancer Immunol Immunother. 2014;63(3):259–71. https://doi.org/10.1007/s00262-013-1506-7

33

Theelen WS, de Jong MC, Baas P. Synergizing systemic responses by combining immunotherapy with radiotherapy in metastatic non‐small cell lung cancer: the potential of the abscopal effect. Lung Cancer. 2020;142:106–13. https://doi.org/10.1016/j.lungcan.2020.02.015

34

Britschgi C, Riesterer O, Burger IA, Guckenberger M, Curioni‐Fontecedro A. Report of an abscopal effect induced by stereotactic body radiotherapy and nivolumab in a patient with metastatic non‐small cell lung cancer. Radiat Oncol. 2018;13(1):102. https://doi.org/10.1186/s13014-018-1049-3

35

Pike LRG, Bang A, Ott P, Balboni T, Taylor A, Catalano P, et al. Radiation and PD‐1 inhibition: favorable outcomes after brain‐directed radiation. Radiother Oncol. 2017;124(1):98–103. https://doi.org/10.1016/j.radonc.2017.06.006

36

Golden EB, Chhabra A, Chachoua A, Adams S, Donach M, Fenton‐Kerimian M, et al. Local radiotherapy and granulocyte–macrophage colony‐stimulating factor to generate abscopal responses in patients with metastatic solid tumours: a proof‐of‐principle trial. Lancet Oncol. 2015;16(7):795–803. https://doi.org/10.1016/S1470-2045(15)00054-6

37

Golden EB, Demaria S, Schiff PB, Chachoua A, Formenti SC. An abscopal response to radiation and ipilimumab in a patient with metastatic non‐small cell lung cancer. Cancer Immunol Res. 2013;1(6):365–72. https://doi.org/10.1158/2326-6066.CIR-13-0115

38

Vanpouille‐Box C, Alard A, Aryankalayil MJ, Sarfraz Y, Diamond JM, Schneider RJ, et al. DNA exonuclease Trex1 regulates radiotherapy‐induced tumour immunogenicity. Nat Commun. 2017;8:15618. https://doi.org/10.1038/ncomms15618

39

Strigari L, Mancuso M, Ubertini V, Soriani A, Giardullo P, Benassi M, et al. Abscopal effect of radiation therapy: interplay between radiation dose and p53 status. Int J Radiat Biol. 2014;90(3):248–55. https://doi.org/10.3109/09553002.2014.874608

40

Camphausen K, O'Reilly M, Menard C, Sproull M, Beecken W, Folkman J, et al. The radiation abscopal anti‐tumor effect is mediated through p53. Int J Radiat Oncol Biol Phys. 2002;54(2):226. https://doi.org/10.1016/s0360-3016(02)03449-1

41

Katayama K, Tamiya A, Koba T, Fukuda S, Atagi S. An abscopal response to radiation therapy in a patient with metastatic non‐small cell lung cancer: a case report. J Cancer Sci Ther. 2017;9(2):365–7. https://doi.org/10.4172/1948-5956.1000443

42

Togitani K, Asagiri T, Iguchi M, Igawa T, Yoshino T, Kojima K. Systemic abscopal effect of low‐dose radiotherapy (2 Gy × 2) against palatine tonsil follicular lymphoma. Intern Med. 2022;61(20):3107–10. https://doi.org/10.2169/internalmedicine.8968-21

43

Donohoe C, Senge MO, Arnaut LG, Gomes‐da‐Silva LC. Cell death in photodynamic therapy: from oxidative stress to anti‐tumor immunity. Biochim Biophys Acta. 2019;1872(2):188308. https://doi.org/10.1016/j.bbcan.2019.07.003

44

Felsher DW. Cancer revoked: oncogenes as therapeutic targets. Nat Rev Cancer. 2003;3(5):375–9. https://doi.org/10.1038/nrc1070

45

Vrouenraets MB, Visser GW, Snow GB, van Dongen GA. Basic principles, applications in oncology and improved selectivity of photodynamic therapy. Anticancer Res. 2003 Jan–Feb;23(1B):505–22.

46

Gudgin Dickson EF, Goyan RL, Pottier RH. New directions in photodynamic therapy. Cell Mol Biol. 2002;48(8):939–54.

47

Dolmans DE, Kadambi A, Hill JS, Flores KR, Gerber JN, Walker JP, et al. Targeting tumor vasculature and cancer cells in orthotopic breast tumor by fractionated photosensitizer dosing photodynamic therapy. Cancer Res. 2002;62(15):4289–94.

48

Dabrowski JM, Arnaut LG. Photodynamic therapy (PDT) of cancer: from local to systemic treatment. Photochem Photobiol Sci. 2015;14(10):1765–80. https://doi.org/10.1039/c5pp00132c

49

Chilakamarthi U, Giribabu L. Photodynamic therapy: past, present and future. Chem Rec. 2017;17(8):775–802. https://doi.org/10.1002/tcr.201600121

50

Garg AD, Agostinis P. ER stress, autophagy and immunogenic cell death in photodynamic therapy‐induced anti‐cancer immune responses. Photochem Photobiol Sci. 2014;13(3):474–87. https://doi.org/10.1039/c3pp50333j

51

Rodrigues MC, Vieira LG, Horst FH, de Araújo EC, Ganassin R, Merker C, et al. Photodynamic therapy mediated by aluminium‐phthalocyanine nanoemulsion eliminates primary tumors and pulmonary metastases in a murine 4T1 breast adenocarcinoma model. J Photochem Photobiol B. 2020;204:111808. https://doi.org/10.1016/j.jphotobiol.2020.111808

52

Zhou S, Zhen Z, Paschall AV, Xue L, Yang X, Bebin Blackwell AG, et al. FAP‐targeted photodynamic therapy mediated by ferritin nanoparticles elicits an immune response against cancer cells and cancer associated fibroblasts. Adv Funct Mater. 2021;31(7):2007017. https://doi.org/10.1002/adfm.202007017

53

Xie Q, Li Z, Liu Y, Zhang D, Su M, Niitsu H, et al. Translocator protein‐targeted photodynamic therapy for direct and abscopal immunogenic cell death in colorectal cancer. Acta Biomater. 2021;134:716–29. https://doi.org/10.1016/j.actbio.2021.07.052

54

Yakkala C, Chiang CLL, Kandalaft L, Denys A, Duran R. Cryoablation and immunotherapy: an enthralling synergy to confront the tumors. Front Immunol. 2019;10:2283. https://doi.org/10.3389/fimmu.2019.02283

55

Hu K. Advances in Clinical Application of Cryoablation Therapy for hepatocellular carcinoma and metastatic liver tumor. J Clin Gastroenterol. 2014;48(10):830–6. https://doi.org/10.1097/MCG.0000000000000201

56

Abdo J, Cornell DL, Mittal SK, Agrawal DK. Immunotherapy plus cryotherapy: potential augmented abscopal effect for advanced cancers. Front Oncol. 2018;8:85. https://doi.org/10.3389/fonc.2018.00085

57

Bastianpillai C, Petrides N, Shah T, Guillaumier S, Ahmed HU, Arya M. Harnessing the immunomodulatory effect of thermal and non‐thermal ablative therapies for cancer treatment. Tumor Biol. 2015;36(12):9137–46. https://doi.org/10.1007/s13277-015-4126-3

58

Soanes WA, Gonder MJ, Ablin RJ. A possible immuno‐cryothermic response in prostatic cancer. Clin Radiol. 1970;21(3):253–5. https://doi.org/10.1016/S0009-9260(70)80036-8

59

Bagley DH, Faraci RP, Marrone JC, Beazley RM. Lymphocyte mediated cytotoxicity after cryosurgery of a murine sarcoma. J Surg Res. 1974;17(6):404–6. https://doi.org/10.1016/0022-4804(74)90151-6

60

Helpap B, Grouls V, Lange O, Breining H, Lymberopoulos S. Morphologic and cell kinetic investigations of the spleen after repeated in situ freezing of liver and kidney. Pathol Res Pract. 1979;164(2):167–77. https://doi.org/10.1016/S0344-0338(79)80021-7

61

Ablin RJ, Soanes WA, Gonder MJ. Elution of in vivo bound antiprostatic epithelial antibodies following multiple cryotherapy of carcinoma of prostate. Urology. 1973;2(3):276–9. https://doi.org/10.1016/0090-4295(73)90463-9

62

Tanaka S. Immunological aspects of cryosurgery in general surgery. Cryobiology. 1982;19(3):247–62. https://doi.org/10.1016/0011-2240(82)90151-1

63

Ablin RJ. Cryosurgery of the rabbit prostate: comparison of the immune response of immature and mature bucks. Cryobiology. 1974;11(5):416–22. https://doi.org/10.1016/0011-2240(74)90108-4

64

Gursel E, Roberts M, Veenema RJ. Regression of prostatic cancer following sequential cryotherapy to the prostate. J Urol. 1972;108(6):928–32. https://doi.org/10.1016/S0022-5347(17)60909-1

65

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(3):376–87. https://doi.org/10.1288/00005537-197303000-00009

66

Blackwood CE, Cooper IS. Response of experimental tumor systems to cryosurgery. Cryobiology. 1972;9(6):508–15. https://doi.org/10.1016/0011-2240(72)90172-1

67

Khan SY, Melkus MW, Rasha F, Castro M, Chu V, Brandi L, et al. Tumor‐infiltrating lymphocytes (TILs) as a biomarker of abscopal effect of cryoablation in breast cancer: a pilot study. Ann Surg Oncol. 2022;29(5):2914–25. https://doi.org/10.1245/s10434-021-11157-w

68

Ko KWS, Chiang JB, Poon WL, Lai E, Garnon J. Abscopal effect after MRI‐guided cryoablation of multifocal chest wall desmoid‐type fibromatosis. Cardiovasc Intervent Radiol. 2020;44(3):509–12. https://doi.org/10.1007/s00270-020-02694-0

69

Yang X, Guo Y, Guo Z, Si T, Xing W, Yu W, et al. Cryoablation inhibition of distant untreated tumors (abscopal effect) is immune mediated. Oncotarget. 2019;10(41):4180–91. https://doi.org/10.18632/oncotarget.24105

70

Liu YE, Zong J, Chen XJ, Zhang R, Ren XC, Guo ZJ, et al. Cryoablation combined with radiotherapy for hepatic malignancy: five case reports. World J Gastrointest Oncol. 2020;12(2):237–47. https://doi.org/10.4251/wjgo.v12.i2.237

71

Yonezawa N, Murakami H, Demura S, Kato S, Miwa S. Abscopal effect of frozen autograft reconstruction combined with an immune checkpoint inhibitor analyzed using a metastatic bone tumor model. Int J Mol Sci. 2021;22(4):1973. https://doi.org/10.3390/ijms22041973

72

Soule E, Bagherpour A, Matteo J. Freezing fort knox: mesenteric carcinoid cryoablation. Gastrointest Tumors. 2017;4(1–2):53–60. https://doi.org/10.1159/000479794

73

Kumar AV, Patterson SG, Plaza MJ. Abscopal effect following cryoablation of breast cancer. J Vasc Interv Radiol. 2019;30(3):466–9. https://doi.org/10.1016/j.jvir.2018.12.004

74

Chokkappan K, Lim MY, Loke SC, Karandikar A, Pua U. Salvage cryoablation of recurrent squamous cell carcinoma for impending airway obstruction with abscopal effect. J Vasc Interv Radiol. 2020;31(11):1939–42. https://doi.org/10.1016/j.jvir.2020.02.007

75

Korman AJ, Peggs KS, Allison JP. Checkpoint blockade in cancer immunotherapy. Adv Immunol. 2006;90:297–339. https://doi.org/10.1016/S0065-2776(06)90008-X

76

Gelao L, Criscitiello C, Esposito A, Goldhirsch A, Curigliano G. Immune checkpoint blockade in cancer treatment: a double‐edged sword cross‐targeting the host as an “innocent bystander”. Toxins. 2014;6(3):914–33. https://doi.org/10.3390/toxins6030914

77

Barker HE, Paget JTE, Khan AA, Harrington KJ. The tumour microenvironment after radiotherapy: mechanisms of resistance and recurrence. Nat Rev Cancer. 2015;15(7):409–25. https://doi.org/10.1038/nrc3958

78

Kingsley DPE. An interesting case of possible abscopal effect in malignant melanoma. Br J Radiol. 1975;48(574):863–6. https://doi.org/10.1259/0007-1285-48-574-863

79

Ehlers G, Fridman M. Abscopal effect of radiation in papillary adenocarcinoma. Br J Radiol. 1973;46(543):220–2. https://doi.org/10.1259/0007-1285-46-543-220

80

Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74. https://doi.org/10.1016/j.cell.2011.02.013

81

Wu A, March L, Zheng X, Huang J, Wang X, Zhao J, et al. Global low back pain prevalence and years lived with disability from 1990 to 2017: estimates from the global burden of disease study 2017. Ann Transl Med. 2020;8(6):299. https://doi.org/10.21037/atm.2020.02.175

82

Bouquet F, Pal A, Pilones KA, Demaria S, Hann B, Akhurst RJ, et al. TGFβ1 inhibition increases the radiosensitivity of breast cancer cells in vitro and promotes tumor control by radiation in vivo. Clin Cancer Res. 2011;17(21):6754–65. https://doi.org/10.1158/1078-0432.CCR-11-0544

83

Wang D, Zhang X, Gao Y, Cui X, Yang Y, Mao W, et al. Research progress and existing problems for abscopal effect. Cancer Manag Res. 2020;12:6695–706. https://doi.org/10.2147/CMAR.S245426

84

Hiniker SM, Chen DS, Reddy S, Chang DT, Jones JC, Mollick JA, et al. A systemic complete response of metastatic melanoma to local radiation and immunotherapy. Transl Oncol. 2012;5(6):404–7. https://doi.org/10.1593/tlo.12280

85

Laoui D, van Overmeire E, de Baetselier P, van Ginderachter JA, Raes G. Functional relationship between tumor‐associated macrophages and macrophage colony‐stimulating factor as contributors to cancer progression. Front Immunol. 2014;5:489. https://doi.org/10.3389/fimmu.2014.00489

86

Reynders K, Illidge T, Siva S, Chang JY, De Ruysscher D. The abscopal effect of local radiotherapy: using immunotherapy to make a rare event clinically relevant. Cancer Treat Rev. 2015;41(6):503–10. https://doi.org/10.1016/j.ctrv.2015.03.011

87

Ashrafizadeh M, Farhood B, Eleojo Musa A, Taeb S, Rezaeyan A, Najafi M. Abscopal effect in radioimmunotherapy. Int Immunopharmacol. 2020;85:106663. https://doi.org/10.1016/j.intimp.2020.106663

88

Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science. 2018;359(6832):1350–5. https://doi.org/10.1126/science.aar4060

89

Qin Q, Nan X, Miller T, Fisher R, Teh B, Pandita S, et al. Complete local and abscopal responses from a combination of radiation and nivolumab in refractory hodgkin's lymphoma. Radiat Res. 2018;190(3):322–9. https://doi.org/10.1667/RR15048.1

90

LaPlant Q, Deselm C, Lockney NA, Hsieh J, Yamada Y. Potential abscopal response to dual checkpoint blockade in RCC after reirradiation using dose‐painting SBRT. Pract Radiat Oncol. 2017;7(6):396–9. https://doi.org/10.1016/j.prro.2017.04.009

91

Dewan MZ, Galloway AE, Kawashima N, Dewyngaert JK, Babb JS, Formenti SC, et al. Fractionated but not single‐dose radiotherapy induces an immune‐mediated abscopal effect when combined with anti‐CTLA‐4 antibody. Clin Cancer Res. 2009;15(17):5379–88. https://doi.org/10.1158/1078-0432.CCR-09-0265

92

Maity A, Mick R, Huang AC, George SM, Farwell MD, Lukens JN, et al. A phase Ⅰ trial of pembrolizumab with hypofractionated radiotherapy in patients with metastatic solid tumours. Br J Cancer. 2018;119(10):1200–7. https://doi.org/10.1038/s41416-018-0281-9

93

Schaue D, Ratikan JA, Iwamoto KS, McBride WH. Maximizing tumor immunity with fractionated radiation. Int J Radiat Oncol Biol Phys. 2012;83(4):1306–10. https://doi.org/10.1016/j.ijrobp.2011.09.049

94

Zhang X, Niedermann G. Abscopal effects with hypofractionated schedules extending into the effector phase of the tumor‐specific T‐cell response. Int J Radiat Oncol Biol Phys. 2018;101(1):63–73. https://doi.org/10.1016/j.ijrobp.2018.01.094

95

Chen M, Qiao G, Hylander BL, Mohammadpour H, Wang XY, Subjeck JR, et al. Adrenergic stress constrains the development of anti‐tumor immunity and abscopal responses following local radiation. Nat Commun. 2020;11(1):1821. https://doi.org/10.1038/s41467-020-15676-0

96

Kang J, Demaria S, Formenti S. Current clinical trials testing the combination of immunotherapy with radiotherapy. J Immunother Cancer. 2016;4:51. https://doi.org/10.1186/s40425-016-0156-7

97

Yin L, Xue J, Li R, Zhou L, Deng L, Chen L, et al. Effect of low‐dose radiation therapy on abscopal responses to hypofractionated radiation therapy and anti‐PD1 in mice and patients with non‐small cell lung cancer. Int J Radiat Oncol Biol Phys. 2020;108(1):212–24. https://doi.org/10.1016/j.ijrobp.2020.05.002

98

Chen D, Barsoumian HB, Yang L, Younes AI, Verma V, Hu Y, et al. SHP‐2 and PD‐L1 inhibition combined with radiotherapy enhances systemic antitumor effects in an anti‐PD‐1‐resistant model of non‐small cell lung cancer. Cancer Immunol Res. 2020;8(7):883–94. https://doi.org/10.1158/2326-6066.CIR-19-0744

99

Yang K, Choi C, Cho H, Ahn WG, Kim SY, Shin SW, et al. Antigen‐capturing mesoporous silica nanoparticles enhance the radiation‐induced abscopal effect in murine hepatocellular carcinoma hepa1‐6 models. Pharmaceutics. 2021;13(11):1811. https://doi.org/10.3390/pharmaceutics13111811

100

Jin L, Duan W, Cai Z, Lim D, Feng Z. Valproic acid triggers radiation‐induced abscopal effect by modulating the unirradiated tumor immune microenvironment in a rat model of breast cancer. J Radiat Res. 2021;62(6):955–65. https://doi.org/10.1093/jrr/rrab037

101

Duan WH, Jin LY, Cai ZC, Lim D, Feng ZH. 2‐Hexyl‐4‐pentylenic acid (HPTA) stimulates the radiotherapy‐induced abscopal effect on distal tumor through polarization of tumor‐associated macrophages. Biomed Environ Sci. 2021;34(9):693–704. https://doi.org/10.3967/bes2021.097

102

Tojo M, Miyato H, Koinuma K, Horie H, Tsukui H, Kimura Y, et al. Metformin combined with local irradiation provokes abscopal effects in a murine rectal cancer model. Sci Rep. 2022;12(1):7290. https://doi.org/10.1038/s41598-022-11236-2

103

Yoon YN, Lee E, Kwon Y‐J, Gim J‐A, Kim T‐J, Kim J‐S. PI3Kδ/γ inhibitor BR101801 extrinsically potentiates effector CD8+ T cell‐dependent antitumor immunity and abscopal effect after local irradiation. J Immunother Cancer. 2022;10(3):e003762. https://doi.org/10.1136/jitc-2021-003762

104

Postow MA, Sidlow R, Hellmann MD. Immune‐related adverse events associated with immune checkpoint blockade. N Engl J Med. 2018;378(2):158–68. https://doi.org/10.1056/nejmra1703481

105

Khoja L, Day D, Wei‐Wu Chen T, Siu LL, Hansen AR. Tumour‐ and class‐specific patterns of immune‐related adverse events of immune checkpoint inhibitors: a systematic review. Ann Oncol. 2017;28(10):2377–85. https://doi.org/10.1093/annonc/mdx286

106

Hao Y, Yasmin‐Karim S, Moreau M, Sinha N, Sajo E, Ngwa W. Enhancing radiotherapy for lung cancer using immunoadjuvants delivered in situ from new design radiotherapy biomaterials: a preclinical study. Phys Med Biol. 2016;61(24):N697–707. https://doi.org/10.1088/1361-6560/61/24/N697

107

Twyman‐Saint Victor C, Rech AJ, Maity A, Rengan R, Pauken KE, Stelekati E, et al. Radiation and dual checkpoint blockade activate non‐redundant immune mechanisms in cancer. Nature. 2015;520(7547):373–7. https://doi.org/10.1038/nature14292

108

Michot JM, Mazeron R, Dercle L, Ammari S, Canova C, Marabelle A, et al. Abscopal effect in a Hodgkin lymphoma patient treated by an anti‐programmed death 1 antibody. Eur J Cancer. 2016;66:91–4. https://doi.org/10.1016/j.ejca.2016.06.017

109

Habets THPM, Oth T, Houben AW, Huijskens MJAJ, Senden‐Gijsbers BLMG, Schnijderberg MCA, et al. Fractionated radiotherapy with 3 × 8 Gy induces systemic anti‐tumour responses and abscopal tumour inhibition without modulating the humoral anti‐tumour response. PLoS One. 2016;11(7):e0159515. https://doi.org/10.1371/journal.pone.0159515

110

Young KH, Baird JR, Savage T, Cottam B, Friedman D, Bambina S, et al. Optimizing timing of immunotherapy improves control of tumors by hypofractionated radiation therapy. PLoS One. 2016;11(6):e0157164. https://doi.org/10.1371/journal.pone.0157164

111

Sharabi A, Kim SS, Kato S, Sanders PD, Patel SP, Sanghvi P, et al. Exceptional response to nivolumab and stereotactic body radiation therapy (SBRT) in neuroendocrine cervical carcinoma with high tumor mutational burden: management considerations from the center for personalized cancer therapy at UC San Diego Moores Cancer Center. Oncologist. 2017;22(6):631–7. https://doi.org/10.1634/theoncologist.2016-0517

112

Sato H, Suzuki Y, Yoshimoto Y, Noda S, Murata K, Takakusagi Y, et al. An abscopal effect in a case of concomitant treatment of locally and peritoneally recurrent gastric cancer using adoptive T‐cell immunotherapy and radiotherapy. Clin Case Rep. 2017;5(4):380–4. https://doi.org/10.1002/ccr3.758

113

Ebner DK, Kamada T, Yamada S. Abscopal effect in recurrent colorectal cancer treated with carbon‐ion radiation therapy: 2 case reports. Adv Radiat Oncol. 2017;2(3):333–8. https://doi.org/10.1016/j.adro.2017.06.001

114

Nasti T, Im S, Kissick HT, Daugherty C, Lawson D, Ahmed R, et al. Effective abscopal response is dependent on optimal radiation dose and sequencing with anti‐PD1 therapy. Int J Radiat Oncol Biol Phys. 2018;102(3):S204. https://doi.org/10.1016/j.ijrobp.2018.07.108

115

Suzuki G, Masui K, Yamazaki H, Takenaka T, Asai S, Taniguchi H, et al. Abscopal effect of high‐dose‐rate brachytherapy on pelvic bone metastases from renal cell carcinoma: a case report. J Contemp Brachyther. 2019;11(5):458–61. https://doi.org/10.5114/jcb.2019.89365

116

Kareff SA, Lischalk JW, Krochmal R, Kim C. Abscopal effect in pulmonary carcinoid tumor following ablative stereotactic body radiation therapy: a case report. J Med Case Rep. 2020;14(1):177. https://doi.org/10.1186/s13256-020-02512-8

117

Chen D, Verma V, Patel RR, Barsoumian HB, Cortez MA, Welsh JW. Absolute lymphocyte count predicts abscopal responses and outcomes in patients receiving combined immunotherapy and radiation therapy: analysis of 3 phase 1/2 trials. Int J Radiat Oncol Biol Phys. 2020;108(1):196–203. https://doi.org/10.1016/j.ijrobp.2020.01.032

118

Agostinis P, Berg K, Cengel KA, Foster TH, Girotti AW, Gollnick SO, et al. Photodynamic therapy of cancer: an update. CA Cancer J Clin. 2011;61(4):250–81. https://doi.org/10.3322/caac.20114

119

Mroz P, Szokalska A, Wu MX, Hamblin MR. Photodynamic therapy of tumors can lead to development of systemic antigen‐specific immune response. PLoS One. 2010;5(12):e15194. https://doi.org/10.1371/journal.pone.0015194

120

Mroz P, Hashmi JT, Huang YY, Lange N, Hamblin MR. Stimulation of anti‐tumor immunity by photodynamic therapy. Expert Rev Clin Immunol. 2011;7(1):75–91. https://doi.org/10.1586/eci.10.81

121

Gallagher WM, Allen LT, O'Shea C, Kenna T, Hall M, Gorman A, et al. A potent nonporphyrin class of photodynamic therapeutic agent: cellular localisation, cytotoxic potential and influence of hypoxia. Br J Cancer. 2005;92(9):1702–10. https://doi.org/10.1038/sj.bjc.6602527

122

Zhou Z, Zhang L, Zhang Z, Liu Z. Advances in photosensitizer‐related design for photodynamic therapy. Asian J Pharm Sci. 2021;16(6):668–86. https://doi.org/10.1016/j.ajps.2020.12.003

123

Huo M, Wang L, Zhang L, Wei C, Chen Y, Shi J. Photosynthetic tumor oxygenation by photosensitizer‐containing Cyanobacteria for enhanced photodynamic therapy. Angew Chem Int Ed. 2020;59(5):1906–13. https://doi.org/10.1002/anie.201912824

124

Jalde SS, Chauhan AK, Lee JH, Chaturvedi PK, Park JS, Kim YW. Synthesis of novel chlorin e6–curcumin conjugates as photosensitizers for photodynamic therapy against pancreatic carcinoma. Eur J Med Chem. 2018;147:66–76. https://doi.org/10.1016/j.ejmech.2018.01.099

125

Luo GF, Chen WH, Hong S, Cheng Q, Qiu WX, Zhang XZ. A self‐transformable pH‐driven membrane‐anchoring photosensitizer for effective photodynamic therapy to inhibit tumor growth and metastasis. Adv Funct Mater. 2017;27:1702122. https://doi.org/10.1002/adfm.201702122

126

Chen D, Yu Q, Huang X, Dai H, Luo T, Shao J, et al. A highly‐efficient type I photosensitizer with robust vascular‐disruption activity for hypoxic‐and‐metastatic tumor specific photodynamic therapy. Small. 2020;16(23):2001059. https://doi.org/10.1002/smll.202001059

127

Liang R, Liu L, He H, Chen Z, Han Z, Luo Z, et al. Oxygen‐boosted immunogenic photodynamic therapy with gold nanocages@manganese dioxide to inhibit tumor growth and metastases. Biomaterials. 2018;177:149–60. https://doi.org/10.1016/j.biomaterials.2018.05.051

128

Hu Y, Huang S, Zhao X, Chang L, Ren X, Mei X, et al. Preparation of photothermal responsive and ROS generative gold nanocages for cancer therapy. Mater Sci Eng C. 2021;125:112098. https://doi.org/10.1016/j.msec.2021.112098

129

Zhu R, He H, Liu Y, Cao D, Yan J, Duan S, et al. Cancer‐selective bioreductive chemotherapy mediated by dual hypoxia‐responsive nanomedicine upon photodynamic therapy‐induced hypoxia aggravation. Biomacromolecules. 2019;20(7):2649–56. https://doi.org/10.1021/acs.biomac.9b00428

130

Yang G, Phua SZF, Lim WQ, Zhang R, Feng L, Liu G, et al. A hypoxia‐responsive albumin‐based nanosystem for deep tumor penetration and excellent therapeutic efficacy. Adv Mater. 2019;31(25):1901513. https://doi.org/10.1002/adma.201901513

131

Li X, Jeon YH, Kwon N, Park JG, Guo T, Kim HR, et al. In vivo‐assembled phthalocyanine/albumin supramolecular complexes combined with a hypoxia‐activated prodrug for enhanced photodynamic immunotherapy of cancer. Biomaterials. 2021;266:120430. https://doi.org/10.1016/j.biomaterials.2020.120430

132

Lan G, Ni K, Xu Z, Veroneau SS, Song Y, Lin W. Nanoscale metal–organic framework overcomes hypoxia for photodynamic therapy primed cancer immunotherapy. J Am Chem Soc. 2018;140:5670–3. https://doi.org/10.1021/jacs.8b01072

133

Lin T, Zhao X, Zhao S, Yu H, Cao W, Chen W, et al. O2‐generating MnO2 nanoparticles for enhanced photodynamic therapy of bladder cancer by ameliorating hypoxia. Theranostics. 2018;8(4):990–1004. https://doi.org/10.7150/thno.22465

134

Lu K, He C, Guo N, Chan C, Ni K, Weichselbaum RR, et al. Chlorin‐based nanoscale metal–organic framework systemically rejects colorectal cancers via synergistic photodynamic therapy and checkpoint blockade immunotherapy. J Am Chem Soc. 2016;138(38):12502–10. https://doi.org/10.1021/jacs.6b06663

135

Chen M, Quan G, Wen T, Yang P, Qin W, Mai H, et al. Cold to hot: binary cooperative microneedle array‐amplified photoimmunotherapy for eliciting antitumor immunity and the abscopal effect. ACS Appl Mater Interfaces. 2020;12(29):32259–69. https://doi.org/10.1021/acsami.0c05090

136

Xing L, Gong JH, Wang Y, Zhu Y, Huang ZJ, Zhao J, et al. Hypoxia alleviation‐triggered enhanced photodynamic therapy in combination with IDO inhibitor for preferable cancer therapy. Biomaterials. 2019;206:170–82. https://doi.org/10.1016/j.biomaterials.2019.03.027

137

Kleinovink JW, Fransen MF, Löwik CW, Ossendorp F. Photodynamic‐immune checkpoint therapy eradicates local and distant tumors by CD8+ T cells. Cancer Immunol Res. 2017;5(10):832–8. https://doi.org/10.1158/2326-6066.CIR-17-0055

138

Hwang HS, Cherukula K, Bang YJ, Vijayan V, Moon MJ, Thiruppathi J, et al. Combination of photodynamic therapy and a flagellin‐adjuvanted cancer vaccine potentiated the anti‐PD‐1‐mediated melanoma suppression. Cells. 2020;9(11):2432. https://doi.org/10.3390/cells9112432

139

Meng L, Cheng Y, Tong X, Gan S, Ding Y, Zhang Y, et al. Tumor oxygenation and hypoxia inducible factor‐1 functional inhibition via a reactive oxygen species responsive nanoplatform for enhancing radiation therapy and abscopal effects. ACS Nano. 2018;12(8):8308–22. https://doi.org/10.1021/acsnano.8b03590

140

Ni K, Luo T, Lan G, Culbert A, Song Y, Wu T, et al. A Nanoscale metal–organic framework to mediate photodynamic therapy and deliver CpG oligodeoxynucleotides to enhance antigen presentation and cancer immunotherapy. Angew Chem Int Ed. 2020;59(3):1108–12. https://doi.org/10.1002/anie.201911429

141

Jin F, Qi J, Liu D, You Y, Shu G, Du Y, et al. Cancer‐cell‐biomimetic upconversion nanoparticles combining chemo‐photodynamic therapy and CD73 blockade for metastatic triple‐negative breast cancer. J Control Rel. 2021;337:90–104. https://doi.org/10.1016/j.jconrel.2021.07.021

142

Kim S, Kim SA, Nam GH, Hong Y, Kim GB, Choi Y, et al. In situ immunogenic clearance induced by a combination of photodynamic therapy and rho‐kinase inhibition sensitizes immune checkpoint blockade response to elicit systemic antitumor immunity against intraocular melanoma and its metastasis. J Immunother Cancer. 2021;9(1):e001481. https://doi.org/10.1136/jitc-2020-001481

143

Xiong W, Qi L, Jiang N, Zhao Q, Chen L, Jiang X, et al. Metformin liposome‐mediated PD‐L1 downregulation for amplifying the photodynamic immunotherapy efficacy. ACS Appl Mater Interfaces. 2021;13(7):8026–41. https://doi.org/10.1021/acsami.0c21743

144

P.S R, Alvi SB, Begum N, Veeresh B, Rengan AK. Self‐assembled fluorosome–polydopamine complex for efficient tumor targeting and commingled photodynamic/photothermal therapy of triple‐negative breast cancer. Biomacromolecules. 2021;22(9):3926–40. https://doi.org/10.1021/acs.biomac.1c00744

145

Li X, Kwon N, Guo T, Liu Z, Yoon J. Innovative strategies for hypoxic‐tumor photodynamic therapy. Angew Chem Int Ed. 2018;57(36):11522–31. https://doi.org/10.1002/anie.201805138

146

Huang Z, Wei G, Zeng Z, Huang Y, Huang L, Shen Y, et al. Enhanced cancer therapy through synergetic photodynamic/immune checkpoint blockade mediated by a liposomal conjugate comprised of porphyrin and IDO inhibitor. Theranostics. 2019;9(19):5542–57. https://doi.org/10.7150/thno.35343

147

Yang G, Xu L, Xu J, Zhang R, Song G, Chao Y, et al. Smart nanoreactors for pH‐responsive tumor homing, mitochondria‐targeting, and enhanced photodynamic‐immunotherapy of cancer. Nano Lett. 2018;18(4):2475–84. https://doi.org/10.1021/acs.nanolett.8b00040

148

O'Shaughnessy MJ, Murray KS, La Rosa SP, Budhu S, Merghoub T, Somma A, et al. Systemic antitumor immunity by PD‐1/PD‐L1 inhibition is potentiated by vascular‐targeted photodynamic therapy of primary tumors. Clin Cancer Res. 2018;24(3):592–9. https://doi.org/10.1158/1078-0432.CCR-17-0186

149

Duan X, Chan C, Guo N, Han W, Weichselbaum RR, Lin W. Photodynamic therapy mediated by nontoxic core‐shell nanoparticles synergizes with immune checkpoint blockade to elicit antitumor immunity and antimetastatic effect on breast cancer. J Am Chem Soc. 2016;138(51):16686–95. https://doi.org/10.1021/jacs.6b09538

150

Yu W, Wang Y, Zhu J, Jin L, Liu B, Xia K, et al. Autophagy inhibitor enhance ZnPc/BSA nanoparticle induced photodynamic therapy by suppressing PD‐L1 expression in osteosarcoma immunotherapy. Biomaterials. 2019;192:128–39. https://doi.org/10.1016/j.biomaterials.2018.11.019

151

Meng Z, Zhou X, Xu J, Han X, Dong Z, Wang H, et al. Light‐triggered in situ gelation to enable robust photodynamic‐Immunotherapy by repeated stimulations. Adv Mater. 2019;31(24):1900927. https://doi.org/10.1002/adma.201900927

152

Zhang R, Zhu Z, Lv H, Li F, Sun S, Li J, et al. Immune checkpoint blockade mediated by a small‐molecule nanoinhibitor targeting the PD‐1/PD‐L1 pathway synergizes with photodynamic therapy to elicit antitumor immunity and antimetastatic effects on breast cancer. Small. 2019;15(49):1903881. https://doi.org/10.1002/smll.201903881

153

Xu J, Xu L, Wang C, Yang R, Zhuang Q, Han X, et al. Near‐infrared‐triggered photodynamic therapy with multitasking upconversion nanoparticles in combination with checkpoint blockade for immunotherapy of colorectal cancer. ACS Nano. 2017;11(4):4463–74. https://doi.org/10.1021/acsnano.7b00715

154

Yu Z, Zhou P, Pan W, Li N, Tang B. A biomimetic nanoreactor for synergistic chemiexcited photodynamic therapy and starvation therapy against tumor metastasis. Nat Commun. 2018;9(1):5044. https://doi.org/10.1038/s41467-018-07197-8

155

Shao Y, Liu B, Di Z, Zhang G, Sun LD, Li L, et al. Engineering of upconverted metal–organic frameworks for near‐infrared light‐triggered combinational photodynamic/chemo‐/immunotherapy against hypoxic tumors. J Am Chem Soc. 2020;142(8):3939–46. https://doi.org/10.1021/jacs.9b12788

156

Deng H, Zhou Z, Yang W, Lin L, Wang S, Niu G, et al. Endoplasmic reticulum targeting to amplify immunogenic cell death for cancer immunotherapy. Nano Lett. 2020;20(3):1928–33. https://doi.org/10.1021/acs.nanolett.9b05210

157

Liu H, Hu Y, Sun Y, Wan C, Zhang Z, Dai X, et al. Co‐delivery of Bee venom melittin and a photosensitizer with an organic–inorganic hybrid nanocarrier for photodynamic therapy and immunotherapy. ACS Nano. 2019;13(11):12638–52. https://doi.org/10.1021/acsnano.9b04181

158

Deng G, Sun Z, Li S, Peng X, Li W, Zhou L, et al. Cell‐membrane immunotherapy based on natural killer cell membrane coated nanoparticles for the effective inhibition of primary and abscopal tumor growth. ACS Nano. 2018;12(12):12096–108. https://doi.org/10.1021/acsnano.8b05292

159

Bloemberg J, Van Riel L, Dodou D, Breedveld P. Focal therapy for localized cancer: a patent review. Expert Rev Med Dev. 2021;18(8):751–69. https://doi.org/10.1080/17434440.2021.1943360

160

Yamashita T, Hayakawa K, Hosokawa M, Kodama T, Inoue N, Tomita K, et al. Enhanced tumor metastases in rats following cryosurgery of primary tumor. Gan. 1982;73(2):222–8.

161
Hayakawa K, Yamashita T, Suzuki K, Tomita K, Hosokawa M, Kodama T, et al. Comparative immunological studies in rats following cryosurgery and surgical excision of 3‐methylcholanthrene‐induced primary autochthonous tumors. Gan. 1982;73(3):462–9. http://europepmc.org/abstract/MED/7129011
162

Miya K, Saji S, Morita T, Niwa H, Sakata K. Experimental study on mechanism of absorption of cryonecrotized tumor antigens. Cryobiology. 1987;24(2):135–9. https://doi.org/10.1016/0011-2240(87)90015-0

163

Shibata T, Yamashita T, Suzuki K, Takeichi N, Micallef M, Hosokawa M, et al. Enhancement of experimental pulmonary metastasis and inhibition of subcutaneously transplanted tumor growth following cryosurgery. Anticancer Res. 1998;18(6A):4443–8.

164

Hanawa S. An experimental study on the induction of anti‐tumor immunological activity after cryosurgery for liver carcinoma, and the effect of concomitant immunotherapy with OK432. Nihon Geka Gakkai Zasshi. 1993;94(1):57–65.

165

Miya K, Saji S, Morita T, Niwa H, Takao H, Kida H, et al. Immunological response of regional lymph nodes after cryosurgery in rats. Nihon Geka Gakkai Zasshi. 1986;87(3):273–7.

166

Viorritto ICB, Nikolov NP, Siegel RM. Autoimmunity versus tolerance: can dying cells tip the balance? Clin Immunol. 2007;122(2):125–34. https://doi.org/10.1016/j.clim.2006.07.012

167

Sabel MS. Cryo‐immunology: a review of the literature and proposed mechanisms for stimulatory versus suppressive immune responses. Cryobiology. 2009;58(1):1–11. https://doi.org/10.1016/j.cryobiol.2008.10.126

168

Scheinecker C, McHugh R, Shevach EM, Germain RN. Constitutive presentation of a natural tissue autoantigen exclusively by dendritic cells in the draining lymph node. J Exp Med. 2002;196(8):1079–90. https://doi.org/10.1084/jem.20020991

169

Peng Y, Martin DA, Kenkel J, Zhang K, Ogden CA, Elkon KB. Innate and adaptive immune response to apoptotic cells. J Autoimmun. 2007;29(4):303–9. https://doi.org/10.1016/j.jaut.2007.07.017

170

Rock KL, Hearn A, Chen C‐J, Shi Y. Natural endogenous adjuvants. Springer Semin Immunopathol. 2005;26:231–46. https://doi.org/10.1007/s00281-004-0173-3

171

Henry F, Boisteau O, Bretaudeau L, Lieubeau B, Meflah K, Grégoire M. Antigen‐presenting cells that phagocytose apoptotic tumor‐derived cells are potent tumor vaccines. Cancer Res. 1999;59(14):3329–32.

172

Scheffer SR, Nave H, Korangy F, Schlote K, Pabst R, Jaffee EM, et al. Apoptotic, but not necrotic, tumor cell vaccines induce a potent immune response in vivo. Int J Cancer. 2003;103(2):205–11. https://doi.org/10.1002/ijc.10777

173

Abdo J, Cornell DL, Mittal SK, Agrawal DK. Immunotherapy plus cryotherapy: potential augmented abscopal effect for advanced cancers. Front Oncol. 2018;8:85. https://doi.org/10.3389/fonc.2018.00085

174

Li F, Guo Z, Yu H, Zhang X, Si T, Liu C, et al. Anti‐tumor immunological response induced by cryoablation and anti‐CTLA‐4 antibody in an in vivo RM‐1 cell prostate cancer murine model. Neoplasma. 2014;61(6):659–71. https://doi.org/10.4149/neo_2014_081

175

Zhu B, Liu Y, Li J, Diao L, Shao L, Han‐Zhang H, et al. Exceptional response of cryoablation followed by pembrolizumab in a patient with metastatic cervical carcinosarcoma with high tumor mutational burden: a case report. Oncologist. 2020;25(1):15–8. https://doi.org/10.1634/theoncologist.2019-0739

176

Doshi A, Zhou M, Bui N, Wang DS, Ganjoo K, Hwang GL. Safety and feasibility of cryoablation during immunotherapy in patients with metastatic soft tissue sarcoma. J Vasc Interv Radiol. 2021;32(12):1688–94. https://doi.org/10.1016/j.jvir.2021.08.017

Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 11 July 2022
Accepted: 29 December 2022
Published: 23 February 2023
Issue date: October 2023

Copyright

© 2023 The Authors. Tsinghua University Press.

Acknowledgements

ACKNOWLEDGMENTS

The authors would like to acknowledge the following agency: Figures 13 were created using BioRender.com.

Rights and permissions

This is an open access article under the terms of the Creative Commons Attribution‐NonCommercial‐NoDerivs License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non‐commercial and no modifications or adaptations are made.

Return